1
|
Fauzi AA, Thamrin AMH, Permana AT, Ranuh IGMAR, Hidayati HB, Hamdan M, Wahyuhadi J, Suroto NS, Lestari P, Chandra PS. Comparison of the Administration Route of Stem Cell Therapy for Ischemic Stroke: A Systematic Review and Meta-Analysis of the Clinical Outcomes and Safety. J Clin Med 2023; 12:jcm12072735. [PMID: 37048818 PMCID: PMC10094955 DOI: 10.3390/jcm12072735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/06/2023] [Accepted: 04/04/2023] [Indexed: 04/09/2023] Open
Abstract
Stem cell treatment is emerging as an appealing alternative for stroke patients, but there still needs to be an agreement on the protocols in place, including the route of administration. This systematic review aimed to assess the efficacy and safety of the administration routes of stem cell treatment for ischemic stroke. A systematic review was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. A comprehensive literature search was undertaken using the PubMed, Scopus, and Cochrane databases. A total of 21 publications on stem cell therapy for ischemic stroke were included. Efficacy outcomes were measured using the National Institutes of Health Stroke Scale (NIHSS), the modified Rankin Scale (mRS), and the Barthel index (BI). Intracerebral administration showed a better outcome than other routes, but a greater number of adverse events followed due to its invasiveness. Adverse events were shown to be related to the natural history of stroke not to the treatment. However, further investigation is required, since studies have yet to compare the different administration methods directly.
Collapse
Affiliation(s)
- Asra Al Fauzi
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Ahmad Muslim Hidayat Thamrin
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Andhika Tomy Permana
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - I. G. M. Aswin R. Ranuh
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Hanik Badriyah Hidayati
- Department of Neurology, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Muhammad Hamdan
- Department of Neurology, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Joni Wahyuhadi
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Nur Setiawan Suroto
- Department of Neurosurgery, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Pudji Lestari
- Department of Public Health and Preventive Medicine, Faculty of Medicine, Universitas Airlangga, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Poodipedi Sarat Chandra
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi 110608, India
| |
Collapse
|
2
|
Yabuno S, Yasuhara T, Nagase T, Kawauchi S, Sugahara C, Okazaki Y, Hosomoto K, Sasada S, Sasaki T, Tajiri N, Borlongan CV, Date I. Synergistic therapeutic effects of intracerebral transplantation of human modified bone marrow-derived stromal cells (SB623) and voluntary exercise with running wheel in a rat model of ischemic stroke. Stem Cell Res Ther 2023; 14:10. [PMID: 36691091 PMCID: PMC9872315 DOI: 10.1186/s13287-023-03236-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cell (MSC) transplantation therapy is a promising therapy for stroke patients. In parallel, rehabilitation with physical exercise could ameliorate stroke-induced neurological impairment. In this study, we aimed to clarify whether combination therapy of intracerebral transplantation of human modified bone marrow-derived MSCs, SB623 cells, and voluntary exercise with running wheel (RW) could exert synergistic therapeutic effects on a rat model of ischemic stroke. METHODS Wistar rats received right transient middle cerebral artery occlusion (MCAO). Voluntary exercise (Ex) groups were trained in a cage with RW from day 7 before MCAO. SB623 cells (4.0 × 105 cells/5 μl) were stereotactically injected into the right striatum at day 1 after MCAO. Behavioral tests were performed at day 1, 7, and 14 after MCAO using the modified Neurological Severity Score (mNSS) and cylinder test. Rats were euthanized at day 15 after MCAO for mRNA level evaluation of ischemic infarct area, endogenous neurogenesis, angiogenesis, and expression of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF). The rats were randomly assigned to one of the four groups: vehicle, Ex, SB623, and SB623 + Ex groups. RESULTS SB623 + Ex group achieved significant neurological recovery in mNSS compared to the vehicle group (p < 0.05). The cerebral infarct area of SB623 + Ex group was significantly decreased compared to those in all other groups (p < 0.05). The number of BrdU/Doublecortin (Dcx) double-positive cells in the subventricular zone (SVZ) and the dentate gyrus (DG), the laminin-positive area in the ischemic boundary zone (IBZ), and the mRNA level of BDNF and VEGF in SB623 + Ex group were significantly increased compared to those in all other groups (p < 0.05). CONCLUSIONS This study suggests that combination therapy of intracerebral transplantation SB623 cells and voluntary exercise with RW achieves robust neurological recovery and synergistically promotes endogenous neurogenesis and angiogenesis after cerebral ischemia, possibly through a mechanism involving the up-regulation of BDNF and VEGF.
Collapse
Affiliation(s)
- Satoru Yabuno
- Department of Neurological Surgery, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558 Japan
| | - Takao Yasuhara
- Department of Neurological Surgery, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558 Japan
| | - Takayuki Nagase
- Department of Neurological Surgery, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558 Japan
| | - Satoshi Kawauchi
- Department of Neurological Surgery, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558 Japan
| | - Chiaki Sugahara
- Department of Neurological Surgery, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558 Japan
| | - Yosuke Okazaki
- Department of Neurological Surgery, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558 Japan
| | - Kakeru Hosomoto
- Department of Neurological Surgery, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558 Japan
| | - Susumu Sasada
- Department of Neurological Surgery, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558 Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558 Japan
| | - Naoki Tajiri
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL USA
| | - Isao Date
- Department of Neurological Surgery, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558 Japan
| |
Collapse
|
3
|
Satani N, Parsha K, Davis C, Gee A, Olson SD, Aronowski J, Savitz SI. Peripheral blood monocytes as a therapeutic target for marrow stromal cells in stroke patients. Front Neurol 2022; 13:958579. [PMID: 36277912 PMCID: PMC9580494 DOI: 10.3389/fneur.2022.958579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
Background Systemic administration of marrow stromal cells (MSCs) leads to the release of a broad range of factors mediating recovery in rodent stroke models. The release of these factors could depend on the various cell types within the peripheral blood as they contact systemically administered MSCs. In this study, we assessed the immunomodulatory interactions of MSCs with peripheral blood derived monocytes (Mϕ) collected from acute stroke patients. Methods Peripheral blood from stroke patients was collected at 5–7 days (N = 5) after symptom onset and from age-matched healthy controls (N = 5) using mononuclear cell preparation (CPT) tubes. After processing, plasma and other cellular fractions were removed, and Mϕ were isolated from the mononuclear fraction using CD14 microbeads. Mϕ were then either cultured alone or co-cultured with MSCs in a trans-well cell-culture system. Secretomes were analyzed after 24 h of co-cultures using a MAGPIX reader. Results Our results show that there is a higher release of IFN-γ and IL-10 from monocytes isolated from peripheral blood at day 5–7 after stroke compared with monocytes from healthy controls. In trans-well co-cultures of MSCs and monocytes isolated from stroke patients, we found statistically significant increased levels of IL-4 and MCP-1, and decreased levels of IL-6, IL-1β, and TNF-α. Addition of MSCs to monocytes increased the secretions of Fractalkine, IL-6, and MCP-1, while the secretions of TNF-α decreased, as compared to the secretions from monocytes alone. When MSCs were added to monocytes from stroke patients, they decreased the levels of IL-1β, and increased the levels of IL-10 significantly more as compared to when they were added to monocytes from control patients. Conclusion The systemic circulation of stroke patients may differentially interact with MSCs to release soluble factors integral to their paracrine mechanisms of benefit. Our study finds that the effect of MSCs on Mϕ is different on those derived from stroke patients blood as compared to healthy controls. These findings suggest immunomodulation of peripheral immune cells as a therapeutic target for MSCs in patients with acute stroke.
Collapse
Affiliation(s)
- Nikunj Satani
- Department of Neurology, McGovern Medical School, Institute for Stroke and Cerebrovascular Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
- *Correspondence: Nikunj Satani
| | - Kaushik Parsha
- Department of Neurology, McGovern Medical School, Institute for Stroke and Cerebrovascular Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Courtney Davis
- Department of Neurology, McGovern Medical School, Institute for Stroke and Cerebrovascular Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Adrian Gee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, United States
| | - Scott D. Olson
- Department of Pediatric Surgery, McGovern Medical School at UTHealth, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jaroslaw Aronowski
- Department of Neurology, McGovern Medical School, Institute for Stroke and Cerebrovascular Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sean I. Savitz
- Department of Neurology, McGovern Medical School, Institute for Stroke and Cerebrovascular Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
4
|
Tang W, Lv X, Huang J, Wang B, Lin L, Shen Y, Yao Y. Neuroprotective Effect of Stroke Pretreated Mesenchymal Stem Cells Against Cerebral Ischemia/Reperfusion Injury in Rats. World Neurosurg 2022; 165:e1-e11. [PMID: 33957285 DOI: 10.1016/j.wneu.2021.04.114] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have been shown to enhance neurological recovery after stroke. A rat middle cerebral artery occlusion model was designed to assess neuroprotective effects of stroke pretreated MSCs on cerebral ischemia/reperfusion injury. METHODS MSCs were isolated and cultured in medium with 10% fetal bovine serum, normal control serum, or stroke serum (SS). MSCs were then injected into rats (n = 6 in each group) 1 day after middle cerebral artery occlusion, and feeding continued for 28 days. A battery of behavioral tests, 2,3,5-triphenyltetrazolium chloride staining, hematoxylin-eosin staining, enzyme-linked immunosorbent assay, and terminal deoxynucleotidyl transferase dUTP nick end labeling assay were used to assess neural injury. To detect enhancement of neuronal regeneration and angiogenesis, immunofluorescence and Western blotting were performed to assess expression of trophic factors and growth factors. RESULTS After treatment, behavior of rats improved significantly. Infarction area, brain lesion, and apoptosis cells were significantly decreased in the SS-MSCs group compared with the other groups. SS-MSCs also modulated inflammation by attenuating inflammatory cytokines. Furthermore, the number of neurogenesis-positive cells and expression of trophic factors and growth factors were significantly higher in the SS-MSCs group compared with the others. MSCs cultured with fetal bovine serum and normal control serum showed differences in expression of trophic factors and growth factors, but the results were not as good as with SS-MSCs. CONCLUSIONS Administration of SS-MCSs after reperfusion led to neuroprotection by inducing the recovery process, including improving pathological changes, behavioral improvement, neurogenesis, suppression of apoptosis and inflammation, and angiogenesis.
Collapse
Affiliation(s)
- Wenxue Tang
- Department of Critical Care Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Xin Lv
- Department of Critical Care Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Jinxiu Huang
- Department of Critical Care Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Baiyong Wang
- Department of Critical Care Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Leqing Lin
- Department of Critical Care Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Yueliang Shen
- Department of Pathophysiology, Zhejiang University Medical College, Hangzhou, China
| | - Yanmei Yao
- Department of General Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China.
| |
Collapse
|
5
|
Samal J, Segura T. Injectable biomaterial shuttles for cell therapy in stroke. Brain Res Bull 2021; 176:25-42. [PMID: 34391821 PMCID: PMC8524625 DOI: 10.1016/j.brainresbull.2021.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 07/26/2021] [Accepted: 08/06/2021] [Indexed: 01/01/2023]
Abstract
Ischemic stroke (IS) is the leading cause of disability and contributes to a significant socio-economic cost in the western world. Brain repair strategies investigated in the pre-clinical models include the delivery of drug or cell-based therapeutics; which is hindered by the complex anatomy and functional organization of the brain. Biomaterials can be instrumental in alleviating some of these challenges by providing a structural support, localization, immunomodulation and/or modulating cellular cross-talk in the brain. This review addresses the significance of and challenges associated with cell therapy in an ischemic brain. This is followed by a detailed insight into the biomaterial-based delivery systems which have been designed to provide sustained trophic factor delivery for endogenous repair and to support transplanted cell survival and integration. A biomaterial intervention uses a multifaceted approach in enhancing the survival and engraftment of cells during transplantation and this has driven them as potential candidates for the treatment of IS. The biological processes that are activated as a response to the biomaterials and how to modulate them is one of the key factors contributing to the success of the biomaterial-based therapeutic approach. Future perspectives highlight the need of a combinative approach of merging the material design with disease biology to fabricate effective biomaterial-based intervention of stroke.
Collapse
Affiliation(s)
- Juhi Samal
- Department of Biomedical Engineering, 534 Research Drive, Durham, NC 27708, United States
| | - Tatiana Segura
- Department of Biomedical Engineering, 534 Research Drive, Durham, NC 27708, United States.
| |
Collapse
|
6
|
Strategies to Improve the Efficiency of Transplantation with Mesenchymal Stem Cells for the Treatment of Ischemic Stroke: A Review of Recent Progress. Stem Cells Int 2021; 2021:9929128. [PMID: 34490053 PMCID: PMC8418553 DOI: 10.1155/2021/9929128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 12/11/2022] Open
Abstract
Cerebral ischemia is a common global disease that is characterized by a loss of neurological function and a poor prognosis in many patients. However, only a limited number of treatments are available for this condition at present. Given that the efficacies of these treatments tend to be poor, cerebral ischemia can create a significant burden on patients, families, and society. Mesenchymal stem cell (MSC) transplantation treatment has shown significant potential in animal models of ischemic stroke; however, the specific mechanisms underlying this effect have yet to be elucidated. Furthermore, clinical trials have yet to yield promising results. Consequently, there is an urgent need to identify new methods to improve the efficiency of MSC transplantation as an optimal treatment for ischemic stroke. In this review, we provide an overview of recent scientific reports concerning novel strategies that promote MSC transplantation as an effective therapeutic approach, including physical approaches, chemical agents, traditional Chinese medicines and extracts, and genetic modification. Our analyses showed that two key factors need to be considered if we are to improve the efficacy of MSC transplantation treatments: survival ability and homing ability. We also highlight the importance of other significant mechanisms, including the enhanced activation of MSCs to promote neurogenesis and angiogenesis, and the regulation of permeability in the blood-brain barrier. Further in-depth investigations of the specific mechanisms underlying MSC transplantation treatment will help us to identify effective methods that improve the efficiency of MSC transplantation for ischemic stroke. The development of safer and more effective methods will facilitate the application of MSC transplantation as a promising adjuvant therapy for the treatment of poststroke brain damage.
Collapse
|
7
|
Duan R, Gao Y, He R, Jing L, Li Y, Gong Z, Yao Y, Luan T, Zhang C, Li L, Jia Y. Induced Pluripotent Stem Cells for Ischemic Stroke Treatment. Front Neurosci 2021; 15:628663. [PMID: 34135724 PMCID: PMC8202685 DOI: 10.3389/fnins.2021.628663] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/06/2021] [Indexed: 12/17/2022] Open
Abstract
Ischemic stroke is one of the main central nervous system diseases and is associated with high disability and mortality rates. Recombinant tissue plasminogen activator (rt-PA) and mechanical thrombectomy are the optimal therapies available currently to restore blood flow in patients with stroke; however, their limitations are well recognized. Therefore, new treatments are urgently required to overcome these shortcomings. Recently, stem cell transplantation technology, involving the transplantation of induced pluripotent stem cells (iPSCs), has drawn the interest of neuroscientists and is considered to be a promising alternative for ischemic stroke treatment. iPSCs are a class of cells produced by introducing specific transcription factors into somatic cells, and are similar to embryonic stem cells in biological function. Here, we have reviewed the current applications of stem cells with a focus on iPSC therapy in ischemic stroke, including the neuroprotective mechanisms, development constraints, major challenges to overcome, and clinical prospects. Based on the current state of research, we believe that stem cells, especially iPSCs, will pave the way for future stroke treatment.
Collapse
Affiliation(s)
- Ranran Duan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Gao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruya He
- The International Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijun Jing
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanfei Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhe Gong
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaobing Yao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tingting Luan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaopeng Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Li
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanjie Jia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Davis C, Savitz SI, Satani N. Mesenchymal Stem Cell Derived Extracellular Vesicles for Repairing the Neurovascular Unit after Ischemic Stroke. Cells 2021; 10:cells10040767. [PMID: 33807314 PMCID: PMC8065444 DOI: 10.3390/cells10040767] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is a debilitating disease and one of the leading causes of long-term disability. During the early phase after ischemic stroke, the blood-brain barrier (BBB) exhibits increased permeability and disruption, leading to an influx of immune cells and inflammatory molecules that exacerbate the damage to the brain tissue. Mesenchymal stem cells have been investigated as a promising therapy to improve the recovery after ischemic stroke. The therapeutic effects imparted by MSCs are mostly paracrine. Recently, the role of extracellular vesicles released by these MSCs have been studied as possible carriers of information to the brain. This review focuses on the potential of MSC derived EVs to repair the components of the neurovascular unit (NVU) controlling the BBB, in order to promote overall recovery from stroke. Here, we review the techniques for increasing the effectiveness of MSC-based therapeutics, such as improved homing capabilities, bioengineering protein expression, modified culture conditions, and customizing the contents of EVs. Combining multiple techniques targeting NVU repair may provide the basis for improved future stroke treatment paradigms.
Collapse
|
9
|
Brain morphological and connectivity changes on MRI after stem cell therapy in a rat stroke model. PLoS One 2021; 16:e0246817. [PMID: 33592008 PMCID: PMC7886198 DOI: 10.1371/journal.pone.0246817] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/26/2021] [Indexed: 01/01/2023] Open
Abstract
In animal models of stroke, behavioral assessments could be complemented by a variety of neuroimaging studies to correlate them with recovery and better understand mechanisms of improvement after stem cell therapy. We evaluated morphological and connectivity changes after treatment with human mesenchymal stem cells (hMSCs) in a rat stroke model, through quantitative measurement of T2-weighted images and diffusion tensor imaging (DTI). Transient middle cerebral artery occlusion rats randomly received PBS (PBS-only), FBS cultured hMSCs (FBS-hMSCs), or stroke patients’ serum cultured hMSCs (SS-hMSCs). Functional improvement was assessed using a modified neurological severity score (mNSS). Quantitative analyses of T2-weighted ischemic lesion and ventricular volume changes were performed. Brain microstructure/connectivity changes were evaluated in the ischemic recovery area by DTI-derived microstructural indices such as relative fractional anisotropy (rFA), relative axial diffusivity (rAD), and relative radial diffusivity (rRD), and relative fiber density (rFD) analyses. According to mNSS results, the SS-hMSCs group showed the most prominent functional improvement. Infarct lesion volume of the SS-hMSCs group was significantly decreased at 2 weeks when compared to the PBS-only groups, but there were no differences between the FBS-hMSCs and SS-hMSCs groups. Brain atrophy was significantly decreased in the SS-hMSCs group compared to the other groups. In DTI, rFA and rFD values were significantly higher and rRD value was significant lower in the SS-hMSCs group and these microstructure/connectivity changes were correlated with T2-weighted morphological changes. T2-weighted volume alterations (ischemic lesion and brain atrophy), and DTI microstructural indices and rFD changes, were well matched with the results of behavioral assessment. These quantitative MRI measurements could be potential outcome predictors of functional recovery after treatment with stem cells for stroke.
Collapse
|
10
|
Affiliation(s)
- Leonardo Roever
- From the Department of Clinical Research (L.R.), Federal University of Uberlândia, Minas Gerais, Brazil; and Faculty of Medicine and Dentistry (G.C.J.), Department of Medicine, University of Alberta, Canada.
| | - Glen C Jickling
- From the Department of Clinical Research (L.R.), Federal University of Uberlândia, Minas Gerais, Brazil; and Faculty of Medicine and Dentistry (G.C.J.), Department of Medicine, University of Alberta, Canada
| |
Collapse
|
11
|
Chung JW, Chang WH, Bang OY, Moon GJ, Kim SJ, Kim SK, Lee JS, Sohn SI, Kim YH. Efficacy and Safety of Intravenous Mesenchymal Stem Cells for Ischemic Stroke. Neurology 2021; 96:e1012-e1023. [PMID: 33472925 DOI: 10.1212/wnl.0000000000011440] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To test whether autologous modified mesenchymal stem cells (MSCs) improve recovery in patients with chronic major stroke. METHODS In this prospective, open-label, randomized controlled trial with blinded outcome evaluation, patients with severe middle cerebral artery territory infarct within 90 days of symptom onset were assigned, in a 2:1 ratio, to receive preconditioned autologous MSC injections (MSC group) or standard treatment alone (control group). The primary outcome was the score on the modified Rankin Scale (mRS) at 3 months. The secondary outcome was to further demonstrate motor recovery. RESULTS A total of 39 and 15 patients were included in the MSC and control groups, respectively, for the final intention-to-treat analysis. Mean age of patients was 68 (range 28-83) years, and mean interval between stroke onset to randomization was 20.2 (range 5-89) days. Baseline characteristics were not different between groups. There was no significant difference between the groups in the mRS score shift at 3 months (p = 0.732). However, secondary analyses showed significant improvements in lower extremity motor function in the MSC group compared to the control group (change in the leg score of the Motricity Index, p = 0.023), which was notable among patients with low predicted recovery potential. There were no serious treatment-related adverse events. CONCLUSIONS IV application of preconditioned, autologous MSCs with autologous serum was feasible and safe in patients with chronic major stroke. MSC treatment was not associated with improvements in the 3-month mRS score, but we did observe leg motor improvement in detailed functional analyses. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that autologous MSCs do not improve 90-day outcomes in patients with chronic stroke. CLINICALTRIALSGOV IDENTIFIER NCT01716481.
Collapse
Affiliation(s)
- Jong-Won Chung
- From the Department of Neurology (J.-W.C., O.Y.B., S.J.K.), Samsung Medical Center, Sungkyunkwan University; Translational and Stem Cell Research Laboratory on Stroke (J.-W.C., O.Y.B., G.J.M.) and Stem Cell and Regenerative Medicine Institute (G.J.M.), Samsung Medical Center; Department of Physical and Rehabilitation Medicine (W.H.C., Y.-H.K.), Center for Prevention and Rehabilitation, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul; School of Life Sciences (G.J.M.), BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu; Department of Neurology (S.-K.K.), Gyeongsang National University School of Medicine, Jinju; Department of Neurology (J.S.L.), Ajou University Hospital, School of Medicine, Suwon; and Department of Neurology (S.-I.S.), Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, South Korea. Dr. Moon is currently affiliated with the Stem Cell Center, Asan Institute for Life Science and the Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Won Hyuk Chang
- From the Department of Neurology (J.-W.C., O.Y.B., S.J.K.), Samsung Medical Center, Sungkyunkwan University; Translational and Stem Cell Research Laboratory on Stroke (J.-W.C., O.Y.B., G.J.M.) and Stem Cell and Regenerative Medicine Institute (G.J.M.), Samsung Medical Center; Department of Physical and Rehabilitation Medicine (W.H.C., Y.-H.K.), Center for Prevention and Rehabilitation, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul; School of Life Sciences (G.J.M.), BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu; Department of Neurology (S.-K.K.), Gyeongsang National University School of Medicine, Jinju; Department of Neurology (J.S.L.), Ajou University Hospital, School of Medicine, Suwon; and Department of Neurology (S.-I.S.), Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, South Korea. Dr. Moon is currently affiliated with the Stem Cell Center, Asan Institute for Life Science and the Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Oh Young Bang
- From the Department of Neurology (J.-W.C., O.Y.B., S.J.K.), Samsung Medical Center, Sungkyunkwan University; Translational and Stem Cell Research Laboratory on Stroke (J.-W.C., O.Y.B., G.J.M.) and Stem Cell and Regenerative Medicine Institute (G.J.M.), Samsung Medical Center; Department of Physical and Rehabilitation Medicine (W.H.C., Y.-H.K.), Center for Prevention and Rehabilitation, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul; School of Life Sciences (G.J.M.), BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu; Department of Neurology (S.-K.K.), Gyeongsang National University School of Medicine, Jinju; Department of Neurology (J.S.L.), Ajou University Hospital, School of Medicine, Suwon; and Department of Neurology (S.-I.S.), Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, South Korea. Dr. Moon is currently affiliated with the Stem Cell Center, Asan Institute for Life Science and the Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Gyeong Joon Moon
- From the Department of Neurology (J.-W.C., O.Y.B., S.J.K.), Samsung Medical Center, Sungkyunkwan University; Translational and Stem Cell Research Laboratory on Stroke (J.-W.C., O.Y.B., G.J.M.) and Stem Cell and Regenerative Medicine Institute (G.J.M.), Samsung Medical Center; Department of Physical and Rehabilitation Medicine (W.H.C., Y.-H.K.), Center for Prevention and Rehabilitation, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul; School of Life Sciences (G.J.M.), BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu; Department of Neurology (S.-K.K.), Gyeongsang National University School of Medicine, Jinju; Department of Neurology (J.S.L.), Ajou University Hospital, School of Medicine, Suwon; and Department of Neurology (S.-I.S.), Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, South Korea. Dr. Moon is currently affiliated with the Stem Cell Center, Asan Institute for Life Science and the Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Suk Jae Kim
- From the Department of Neurology (J.-W.C., O.Y.B., S.J.K.), Samsung Medical Center, Sungkyunkwan University; Translational and Stem Cell Research Laboratory on Stroke (J.-W.C., O.Y.B., G.J.M.) and Stem Cell and Regenerative Medicine Institute (G.J.M.), Samsung Medical Center; Department of Physical and Rehabilitation Medicine (W.H.C., Y.-H.K.), Center for Prevention and Rehabilitation, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul; School of Life Sciences (G.J.M.), BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu; Department of Neurology (S.-K.K.), Gyeongsang National University School of Medicine, Jinju; Department of Neurology (J.S.L.), Ajou University Hospital, School of Medicine, Suwon; and Department of Neurology (S.-I.S.), Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, South Korea. Dr. Moon is currently affiliated with the Stem Cell Center, Asan Institute for Life Science and the Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Soo-Kyoung Kim
- From the Department of Neurology (J.-W.C., O.Y.B., S.J.K.), Samsung Medical Center, Sungkyunkwan University; Translational and Stem Cell Research Laboratory on Stroke (J.-W.C., O.Y.B., G.J.M.) and Stem Cell and Regenerative Medicine Institute (G.J.M.), Samsung Medical Center; Department of Physical and Rehabilitation Medicine (W.H.C., Y.-H.K.), Center for Prevention and Rehabilitation, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul; School of Life Sciences (G.J.M.), BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu; Department of Neurology (S.-K.K.), Gyeongsang National University School of Medicine, Jinju; Department of Neurology (J.S.L.), Ajou University Hospital, School of Medicine, Suwon; and Department of Neurology (S.-I.S.), Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, South Korea. Dr. Moon is currently affiliated with the Stem Cell Center, Asan Institute for Life Science and the Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jin Soo Lee
- From the Department of Neurology (J.-W.C., O.Y.B., S.J.K.), Samsung Medical Center, Sungkyunkwan University; Translational and Stem Cell Research Laboratory on Stroke (J.-W.C., O.Y.B., G.J.M.) and Stem Cell and Regenerative Medicine Institute (G.J.M.), Samsung Medical Center; Department of Physical and Rehabilitation Medicine (W.H.C., Y.-H.K.), Center for Prevention and Rehabilitation, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul; School of Life Sciences (G.J.M.), BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu; Department of Neurology (S.-K.K.), Gyeongsang National University School of Medicine, Jinju; Department of Neurology (J.S.L.), Ajou University Hospital, School of Medicine, Suwon; and Department of Neurology (S.-I.S.), Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, South Korea. Dr. Moon is currently affiliated with the Stem Cell Center, Asan Institute for Life Science and the Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sung-Il Sohn
- From the Department of Neurology (J.-W.C., O.Y.B., S.J.K.), Samsung Medical Center, Sungkyunkwan University; Translational and Stem Cell Research Laboratory on Stroke (J.-W.C., O.Y.B., G.J.M.) and Stem Cell and Regenerative Medicine Institute (G.J.M.), Samsung Medical Center; Department of Physical and Rehabilitation Medicine (W.H.C., Y.-H.K.), Center for Prevention and Rehabilitation, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul; School of Life Sciences (G.J.M.), BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu; Department of Neurology (S.-K.K.), Gyeongsang National University School of Medicine, Jinju; Department of Neurology (J.S.L.), Ajou University Hospital, School of Medicine, Suwon; and Department of Neurology (S.-I.S.), Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, South Korea. Dr. Moon is currently affiliated with the Stem Cell Center, Asan Institute for Life Science and the Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yun-Hee Kim
- From the Department of Neurology (J.-W.C., O.Y.B., S.J.K.), Samsung Medical Center, Sungkyunkwan University; Translational and Stem Cell Research Laboratory on Stroke (J.-W.C., O.Y.B., G.J.M.) and Stem Cell and Regenerative Medicine Institute (G.J.M.), Samsung Medical Center; Department of Physical and Rehabilitation Medicine (W.H.C., Y.-H.K.), Center for Prevention and Rehabilitation, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul; School of Life Sciences (G.J.M.), BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu; Department of Neurology (S.-K.K.), Gyeongsang National University School of Medicine, Jinju; Department of Neurology (J.S.L.), Ajou University Hospital, School of Medicine, Suwon; and Department of Neurology (S.-I.S.), Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, South Korea. Dr. Moon is currently affiliated with the Stem Cell Center, Asan Institute for Life Science and the Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | | |
Collapse
|
12
|
Mesenchymal stem cell therapy for ischemic stroke: A look into treatment mechanism and therapeutic potential. J Neurol 2020; 268:4095-4107. [DOI: 10.1007/s00415-020-10138-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022]
|
13
|
Mangin G, Kubis N. Cell Therapy for Ischemic Stroke: How to Turn a Promising Preclinical Research into a Successful Clinical Story. Stem Cell Rev Rep 2020; 15:176-193. [PMID: 30443706 DOI: 10.1007/s12015-018-9864-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stroke is a major public health issue with limited treatment. The pharmacologically or mechanically removing of the clot is accessible to less than 10% of the patients. Stem cell therapy is a promising alternative strategy since it increases the therapeutic time window but many issues remain unsolved. To avoid a new dramatic failure when translating experimental data on the bedside, this review aims to highlight the indispensable checkpoints to make a successful clinical trial based on the current preclinical literature. The large panel of progenitors/ stem cells at the researcher's disposal is to be used wisely, regarding the type of cells, the source of cells, the route of delivery, the time window, since it will directly affect the outcome. Mechanisms are still incompletely understood, although recent studies have focused on the inflammation modulation of most cells types.
Collapse
Affiliation(s)
| | - Nathalie Kubis
- INSERM U965, F-75475, Paris, France. .,Sorbonne Paris Cité, Université Paris Diderot, F-75475, Paris, France. .,Service de Physiologie Clinique-Explorations Fonctionnelles, AP-HP, Hôpital Lariboisière, 2 rue Ambroise Paré, F-75475, Paris, France.
| |
Collapse
|
14
|
Song B, Wang XX, Yang HY, Kong LT, Sun HY. Temperature-sensitive bone mesenchymal stem cells combined with mild hypothermia reduces neurological deficit in rats of severe traumatic brain injury. Brain Inj 2020; 34:975-982. [PMID: 32362186 DOI: 10.1080/02699052.2020.1753112] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND To explore the combined influences of temperature-sensitive bone mesenchymal stem cells (tsBMSCs) and mild hypothermia (MH) on neurological function and glucose metabolism in rats with severe traumatic brain injury (TBI). METHODS SD rats were randomly divided into sham, TBI, TBI + MH, TBI + BMSCs and TBI + MH +tsBMSCs groups. Then, the brain water content, serum-specific proteins (S100β, NSE, LDH, and CK), and blood glucose at different time points were measured. Furthermore, GLUT-3 expression was detected by Western blotting, and apoptotic rate was determined by TUNEL staining. RESULTS After TBI rat establishment, the brain injury resulted in significant increases in mNSS scores and brain water content, and upregulations in serum levels of S100β, NSE, LDH and CK, and blood glucose, with the elevated cell apoptotic rate in the injured cortex. However, these changes were reversed by MH alone, BMSCs alone, or combination treatment of MH and tsBMSCs in varying degrees, and the combination treatment was superior to the treatment with BMSCs or MH alone. CONCLUSION Combination therapy of tsBMSCs and MH can reduce the neuronal apoptosis in severe TBI rats, with the suppression of serum biomarkers and hyperglycemia, contributing to the recovery of neurological functions. ABBREVIATIONS tsBMSCs: temperature-sensitive bone mesenchymal stem cells; MH: mild hypothermia; TBI: traumatic brain injury; mNSS: modified Neurological Severity Score.
Collapse
Affiliation(s)
- Bo Song
- Department of Emergency, YanTaiShan Hospital , YanTai, Shandong, China
| | - Xin-Xiang Wang
- Department of Laboratory, Yantai Chefoo Area Directly Subordinate Organ Hospital , YanTai, Shandong, China
| | - Hai-Yan Yang
- Department of Emergency, YanTaiShan Hospital , YanTai, Shandong, China
| | - Ling-Ting Kong
- Department of Emergency, YanTaiShan Hospital , YanTai, Shandong, China
| | - Hong-Yan Sun
- Department of Endocrinology, YanTaiShan Hospital , YanTai, Shandong, China
| |
Collapse
|
15
|
Liu S, Guo R, Hou X, Zhang Y, Jiang X, Wang T, Wu X, Xu K, Pan X, Qiao L. Adipose-tissue derived porcine mesenchymal stem cells efficiently ameliorate CCl 4-induced acute liver failure in mice. Cytotechnology 2020; 72:327-341. [PMID: 32335812 DOI: 10.1007/s10616-020-00370-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 01/09/2020] [Indexed: 01/16/2023] Open
Abstract
Adipose tissue derived mesenchymal stem cells (ADMSCs) may be an attractive therapeutic source for acute liver injury because of their high accessibility and non-invasiveness. Here, we investigated the therapeutic potentials of porcine ADMSCs for acute liver failure (ALF). The morphology, differentiation potential, expression patterns of cell surface markers and liver-specific genes were compared between the ADMSCs derived from the pigs with or without ALF. For therapeutic studies, the expanded porcine ADMSCs from either ALF pig (ALF-ADMSCs) or healthy control pig (Nor-ADMSCs) of passage 3 were transplanted into CCl4-induced ALF mice, and the liver histology and functional tests were performed at days 1, 7, 14, and 21 after cell transplantation. ALF-ADMSCs expressed higher mRNA level of hepatic growth factor (HGF) than the Nor-ADMSCs. Both ALF-ADMSCs and Nor-ADMSCs improved liver histology, functions, and mouse survival rate. Higher level of porcine hepatocyte-specific genes was seen in the livers of ALF-ADMSCs transplanted mice as compared to the Nor-ADMSCs transplanted mice. In particular, ALF-ADMSCs transplanted mice expressed significantly higher level of albumin and cytokeratin 18 in the liver tissues as compared to the Nor-ADMSCs transplanted mice. ALF-ADMSCs might be superior to Nor-ADMSCs in the treatment of ALF as the former possesses stronger hepatic differentiation potential.
Collapse
Affiliation(s)
- Shourong Liu
- Department of Liver Diseases, Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310014, China.
| | - Ruihong Guo
- The Fourth Clinical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaoli Hou
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yue Zhang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiawei Jiang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Tiantian Wang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaoyu Wu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Keyang Xu
- The Fourth Clinical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaoping Pan
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
16
|
Venkat P, Cui C, Chopp M, Zacharek A, Wang F, Landschoot-Ward J, Shen Y, Chen J. MiR-126 Mediates Brain Endothelial Cell Exosome Treatment-Induced Neurorestorative Effects After Stroke in Type 2 Diabetes Mellitus Mice. Stroke 2019; 50:2865-2874. [PMID: 31394992 DOI: 10.1161/strokeaha.119.025371] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background and Purpose- Stroke patients with type 2 diabetes mellitus (T2DM) exhibit increased vascular and white matter damage and have worse prognosis compared with nondiabetic stroke patients. We investigated the neurorestorative effects of exosomes derived from mouse brain endothelial cells (EC-Exo) as treatment for stroke in T2DM mice and investigated the role of miR-126 in mediating EC-Exo-derived therapeutic benefits in T2DM-stroke mice. Methods- Adult, male BKS.Cg-m+/+Leprdb/J (T2DM) mice were subjected to photothrombotic stroke model. T2DM mice were intravenously injected at 3 days after stroke with (1) PBS; (2) liposome mimic (vehicle control, 3×1010); (3) EC-Exo (3×1010); (4) knockdown of miR-126 in EC-Exo (miR-126-/- EC-Exo, 3×1010). Behavioral and cognitive tests were performed, and mice were sacrificed at 28 days after stroke. Results- Compared with non-DM stroke mice, T2DM-stroke mice exhibit significantly decreased serum and brain tissue miR-126 expression. Endothelial cells and EC-Exo contain high levels of miR-126 compared with other cell types or exosomes derived from other types of cells, respectively (smooth muscle cells, astrocytes, and marrow stromal cells). Compared with PBS or liposome mimic treatment, EC-Exo treatment of T2DM-stroke mice significantly improves neurological and cognitive function, increases axon density, myelin density, vascular density, arterial diameter, as well as induces M2 macrophage polarization in the ischemic boundary zone. MiR-126-/- EC-Exo treatment significantly decreases miR-126 expression in serum and brain, as well as attentuates EC-Exo treatment-induced functional improvement and does not significantly increase axon and myelin density, vascular density, arterial diameter or induce M2 macrophage polarization in T2DM-stroke mice. In vitro, EC-Exo treatment significantly increases primary cortical neuron axonal outgrowth and increases endothelial capillary tube formation whereas miR-126-/- EC-Exo attentuates EC-Exo induced capillary tube formation and axonal outgrowth. Conclusions- EC-Exo treatment of stroke promotes neurorestorative effects in T2DM mice. MiR-126 may mediate EC-Exo-induced neurorestorative effects in T2DM mice. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Poornima Venkat
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., C.C., M.C., A.Z., F.W., J.L.-W., Y.S., J.C.)
| | - Chengcheng Cui
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., C.C., M.C., A.Z., F.W., J.L.-W., Y.S., J.C.)
| | - Michael Chopp
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., C.C., M.C., A.Z., F.W., J.L.-W., Y.S., J.C.).,Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Alex Zacharek
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., C.C., M.C., A.Z., F.W., J.L.-W., Y.S., J.C.)
| | - Fengjie Wang
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., C.C., M.C., A.Z., F.W., J.L.-W., Y.S., J.C.)
| | - Julie Landschoot-Ward
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., C.C., M.C., A.Z., F.W., J.L.-W., Y.S., J.C.)
| | - Yi Shen
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., C.C., M.C., A.Z., F.W., J.L.-W., Y.S., J.C.)
| | - Jieli Chen
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., C.C., M.C., A.Z., F.W., J.L.-W., Y.S., J.C.)
| |
Collapse
|
17
|
Yang C, Yang Y, Ma L, Zhang GX, Shi FD, Yan Y, Chang G. Study of the cytological features of bone marrow mesenchymal stem cells from patients with neuromyelitis optica. Int J Mol Med 2019; 43:1395-1405. [PMID: 30628649 PMCID: PMC6365084 DOI: 10.3892/ijmm.2019.4056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/04/2019] [Indexed: 12/22/2022] Open
Abstract
Neuromyelitis optica (NMO) is a refractory autoimmune inflammatory disease of the central nervous system without an effective cure. Autologous bone marrow-derived mesenchymal stem cells (BM-MSCs) are considered to be promising therapeutic agents for this disease due to their potential regenerative, immune regulatory and neurotrophic effects. However, little is known about the cytological features of BM-MSCs from patients with NMO, which may influence any therapeutic effects. The present study aimed to compare the proliferation, differentiation and senescence of BM-MSCs from patients with NMO with that of age- and sex-matched healthy subjects. It was revealed that there were no significant differences in terms of cell morphology or differentiation capacities in the BM-MSCs from the patients with NMO. However, in comparison with healthy controls, BM-MSCs derived from the Patients with NMO exhibited a decreased proliferation rate, in addition to a decreased expression of several cell cycle-promoting and proliferation-associated genes. Furthermore, the cell death rate increased in BM-MSCs from patients under normal culture conditions and an assessment of the gene expression profile further confirmed that the BM-MSCs from patients with NMO were more vulnerable to senescence. Platelet-derived growth factor (PDGF), as a major mitotic stimulatory factor for MSCs and a potent therapeutic cytokine in demyelinating disease, was able to overcome the decreased proliferation rate and increased senescence defects in BM-MSCs from the patients with NMO. Taken together, the results from the present study have enabled the proposition of the possibility of combining the application of autologous BM-MSCs and PDGF for refractory and severe patients with NMO in order to elicit improved therapeutic effects, or, at the least, to include PDGF as a necessary and standard growth factor in the current in vitro formula for the culture of NMO patient-derived BM-MSCs.
Collapse
Affiliation(s)
- Chunsheng Yang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yang Yang
- Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin 300052, P.R. China
| | - Li Ma
- Department of Neurosurgery and Neuro‑Oncology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yaping Yan
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Guoqiang Chang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
18
|
Cunningham CJ, Redondo-Castro E, Allan SM. The therapeutic potential of the mesenchymal stem cell secretome in ischaemic stroke. J Cereb Blood Flow Metab 2018; 38:1276-1292. [PMID: 29768965 PMCID: PMC6077926 DOI: 10.1177/0271678x18776802] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) hold great potential as a regenerative therapy for stroke, leading to increased repair and functional recovery in animal models of cerebral ischaemia. While it was initially hypothesised that cell replacement was an important mechanism of action of MSCs, focus has shifted to their paracrine actions or the so called "bystander" effect. MSCs secrete a wide array of growth factors, chemokines, cytokines and extracellular vesicles, commonly referred to as the MSC secretome. There is evidence suggesting the MSC secretome can promote repair through a number of mechanisms including preventing cell apoptosis, modulating the inflammatory response and promoting endogenous repair mechanisms such as angiogenesis and neurogenesis. In this review, we will discuss the in vitro approaches currently being employed to drive the MSC secretome towards a more anti-inflammatory and regenerative phenotype. We will then examine the role of the secretome in promoting repair and improving recovery in preclinical models of cerebral ischaemia.
Collapse
Affiliation(s)
- Catriona J Cunningham
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Elena Redondo-Castro
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
19
|
Moon GJ, Cho YH, Kim DH, Sung JH, Son JP, Kim S, Cha JM, Bang OY. Serum-mediated Activation of Bone Marrow-derived Mesenchymal Stem Cells in Ischemic Stroke Patients: A Novel Preconditioning Method. Cell Transplant 2018; 27:485-500. [PMID: 29774769 PMCID: PMC6038038 DOI: 10.1177/0963689718755404] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Stroke induces complex and dynamic, local and systemic changes including inflammatory
reactions, immune responses, and repair and recovery processes. Mesenchymal stem cells
(MSCs) have been shown to enhance neurological recovery after stroke. We hypothesized that
serum factors play a critical role in the activation of bone marrow (BM) MSCs after stroke
such as by increasing proliferation, paracrine effects, and rejuvenation. Human MSCs
(hMSCs) were grown in fetal bovine serum (FBS), normal healthy control serum (NS), or
stroke patient serum (SS). MSCs cultured in growth medium with 10% SS or NS exhibited
higher proliferation indices than those cultured with FBS (P < 0.01).
FBS-, NS-, and SS-hMSCs showed differences in the expression of trophic factors; vascular
endothelial growth factor, glial cell–derived neurotrophic factor, and fibroblast growth
factor were densely expressed in samples cultured with SS (P < 0.01).
In addition, SS-MSCs revealed different cell cycle– or aging-associated messenger RNA
expression in a later passage, and β-galactosidase staining showed the senescence of MSCs
observed during culture expansion was lower in MSCs cultured with SS than those cultured
with NS or FBS (P < 0.01). Several proteins related to the activity of
receptors, growth factors, and cytokines were more prevalent in the serum of stroke
patients than in that of normal subjects. Neurogenesis and angiogenesis were markedly
increased in rats that had received SS-MSCs (P < 0.05), and these rats
showed significant behavioral improvements (P < 0.01). Our results
indicate that stroke induces a process of recovery via the activation of MSCs. Culture
methods for MSCs using SS obtained during the acute phase of a stroke could constitute a
novel MSC activation method that is feasible and efficient for the neurorestoration of
stroke.
Collapse
Affiliation(s)
- Gyeong Joon Moon
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,2 Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea.,3 School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Buk-gu, Daegu, South Korea
| | - Yeon Hee Cho
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,4 Samsung Biomedical Research Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea
| | - Dong Hee Kim
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,5 Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Jongno-gu, Seoul, South Korea
| | - Ji Hee Sung
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,4 Samsung Biomedical Research Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea
| | - Jeong Pyo Son
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,5 Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Jongno-gu, Seoul, South Korea
| | - Sooyoon Kim
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,4 Samsung Biomedical Research Institute, Samsung Medical Center, Gangnam-gu, Seoul, South Korea
| | - Jae Min Cha
- 6 Medical Device Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Oh Young Bang
- 1 Translational and Stem Cell Research Laboratory on Stroke, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,5 Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Jongno-gu, Seoul, South Korea.,7 Department of Neurology, Samsung Medical Center, Sungkyunkwan University, Jongno-gu, Seoul, South Korea
| |
Collapse
|
20
|
Xue P, Wang M, Yan G. Mesenchymal stem cell transplantation as an effective treatment strategy for ischemic stroke in Asia: a meta-analysis of controlled trials. Ther Clin Risk Manag 2018; 14:909-928. [PMID: 29785117 PMCID: PMC5957058 DOI: 10.2147/tcrm.s161326] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective The aim of this study was to evaluate the efficacy and safety of the mesenchymal stem cell (MSC) therapy in patients with ischemic stroke (IS). Materials and methods Clinical trials involved in this research were searched from PubMed, Web of Science, Cochrane Library, Embase, Wanfang and CNKI database. Therapeutic effects of MSC therapy were assessed according to National Institutes of Health Stroke Scale (NIHSS), Barthel index (BI), Fugl-Meyer Assessment (FMA) and Functional Independence Measure (FIM), and its safety was evaluated based on adverse events. Results This research covered 23 trials including 1,279 IS patients. Based on our analysis, the overall condition of IS patients significantly improved after MSC therapy, indicated by decreased NIHSS and increased BI, FMA and FIM scores. Our analysis also showed that the treatment effects in the MSC transplantation group were superior to those in the control group (routine medication therapy) with statistical significance for NIHSS (1 month after therapy: odds ratio [OR]=-1.92, CI=-3.49 to -0.34, P=0.02; 3 months after therapy: OR=-2.65, CI=-3.40 to -1.90, P<0.00001), BI (1 month after therapy: OR=0.99, CI=0.19-1.79, P=0.02; 6 months after therapy: OR=10.10, CI=3.07-17.14, P=0.005), FMA (3 months after therapy: OR=10.20, CI=3.70-16.70, P=0.002; 6 months after therapy: OR=10.82, CI=6.45-15.18, P<0.00001) and FIM (1 month after therapy: OR=15.61, CI=-0.02 to 31.24, P=0.05; 6 months after therapy: OR=16.56, CI=9.06-24.06, P<0.0001). No serious adverse events were reported during MSC therapy. Conclusion MSC therapy is safe and effective in treating IS by improving the neurological deficits, motor function and daily life quality of patients.
Collapse
Affiliation(s)
- Ping Xue
- Department of Neurology, Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, People's Republic of China
| | - Min Wang
- Department of Neurology, Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, People's Republic of China
| | - Guanhua Yan
- Department of Neurology, Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, People's Republic of China
| |
Collapse
|
21
|
Bang OY, Moon GJ, Kim DH, Lee JH, Kim S, Son JP, Cho YH, Chang WH, Kim YH. Stroke Induces Mesenchymal Stem Cell Migration to Infarcted Brain Areas Via CXCR4 and C-Met Signaling. Transl Stroke Res 2017; 8:10.1007/s12975-017-0538-2. [PMID: 28547726 DOI: 10.1007/s12975-017-0538-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/31/2017] [Accepted: 05/08/2017] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells circulate between organs to repair and maintain tissues. Mesenchymal stem cells cultured with fetal bovine serum have therapeutic effects when intravenously administered after stroke. However, only a small number of mesenchymal stem cells reach the brain. We hypothesized that the serum from stroke patients increases mesenchymal stem cells trophism toward the infarcted brain area. Mesenchymal stem cells were grown in fetal bovine serum, normal serum from normal rats, or stroke serum from ischemic stroke rats. Compared to the fetal bovine serum group, the stroke serum group but not the normal serum group showed significantly greater migration toward the infarcted brain area in the in vitro and in vivo models (p < 0.05). Both C-X-C chemokine receptor type 4 and c-Met expression levels significantly increased in the stroke serum group than the others. The enhanced mesenchymal stem cells migration of the stroke serum group was abolished by inhibition of signaling. Serum levels of chemokines, cytokines, matrix metalloproteinase, and growth factors were higher in stroke serum than in normal serum. Behavioral tests showed a significant improvement in the recovery after stroke in the stroke serum group than the others. Stroke induces mesenchymal stem cells migration to the infarcted brain area via C-X-C chemokine receptor type 4 and c-Met signaling. Culture expansion using the serum from stroke patients could constitute a novel preconditioning method to enhance the therapeutic efficiency of mesenchymal stem cells.
Collapse
Affiliation(s)
- Oh Young Bang
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University, 50 Irwon-dong, Gangnam-gu, Seoul, 135-710, South Korea.
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea.
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, South Korea.
| | - Gyeong Joon Moon
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, South Korea
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea
| | - Dong Hee Kim
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, South Korea
| | - Ji Hyun Lee
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, South Korea
| | - Sooyoon Kim
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, South Korea
| | - Jeong Pyo Son
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, South Korea
| | - Yeon Hee Cho
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, South Korea
| | - Won Hyuk Chang
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| | - Yun-Hee Kim
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| |
Collapse
|
22
|
Li L, Chu L, Fang Y, Yang Y, Qu T, Zhang J, Yin Y, Gu J. Preconditioning of bone marrow-derived mesenchymal stromal cells by tetramethylpyrazine enhances cell migration and improves functional recovery after focal cerebral ischemia in rats. Stem Cell Res Ther 2017; 8:112. [PMID: 28499457 PMCID: PMC5429508 DOI: 10.1186/s13287-017-0565-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/23/2017] [Accepted: 04/26/2017] [Indexed: 12/13/2022] Open
Abstract
Background Transplantation of bone marrow-derived mesenchymal stem cells (BMSCs) is one of the new therapeutic strategies for treating ischemic stroke. However, the relatively poor migratory capacity of BMSCs toward infarcted regions limited the therapeutic potential of this approach. Pharmacological preconditioning can increase the expression of CXC chemokine receptor 4 (CXCR4) in BMSCs and enhance cell migration toward the injury site. In the present study, we investigated whether tetramethylpyrazine (TMP) preconditioning could enhance BMSCs migration to the ischemic brain and improve functional recovery through upregulating CXCR4 expression. Methods BMSCs were identified by flow cytometry analysis. BMSCs migration was evaluated in vitro by transwell migration assay, and CXCR4 expression was measured by quantitative reverse transcription-polymerase chain reaction and western blot analysis. In rats with focal cerebral ischemia, the neurological function was evaluated by the modified neurological severity score, the adhesive removal test and the corner test. The homing BMSCs and angiogenesis were detected by immunofluorescence, and expression of stromal cell-derived factor-1 (SDF-1) and CXCR4 was measured by western blot analysis. Results Flow cytometry analysis demonstrated that BMSCs expressed CD29 and CD90, but not CD34 and CD45. TMP pretreatment dose-dependently induced BMSCs migration and CXCR4 expression in vitro, which was significantly inhibited by AMD3100, a CXCR4 antagonist. In rat stroke models, we found more TMP-preconditioned BMSCs homing toward the infarcted regions than nonpreconditioned cells, leading to improved neurological performance and enhanced angiogenesis. Moreover, TMP-preconditioned BMSCs significantly upregulated the protein expression of SDF-1 and CXCR4 in the ischemic boundary regions. These beneficial effects of TMP preconditioning were blocked by AMD3100. Conclusion TMP preconditioning enhances the migration and homing ability of BMSCs, increases CXCR4 expression, promotes angiogenesis, and improves neurological performance. Therefore, TMP preconditioning may be an effective strategy to improve the therapeutic potency of BMSCs for ischemic stroke due to enhanced BMSCs migration to ischemic regions.
Collapse
Affiliation(s)
- Lin Li
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lisheng Chu
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yan Fang
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yan Yang
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Tiebing Qu
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jianping Zhang
- Department of Anatomy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yuanjun Yin
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jingjing Gu
- Department of Pathology, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
23
|
Bang OY, Kim EH, Cha JM, Moon GJ. Adult Stem Cell Therapy for Stroke: Challenges and Progress. J Stroke 2016; 18:256-266. [PMID: 27733032 PMCID: PMC5066440 DOI: 10.5853/jos.2016.01263] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/15/2016] [Accepted: 09/18/2016] [Indexed: 02/06/2023] Open
Abstract
Stroke is one of the leading causes of death and physical disability among adults. It has been 15 years since clinical trials of stem cell therapy in patients with stroke have been conducted using adult stem cells like mesenchymal stem cells and bone marrow mononuclear cells. Results of randomized controlled trials showed that adult stem cell therapy was safe but its efficacy was modest, underscoring the need for new stem cell therapy strategies. The primary limitations of current stem cell therapies include (a) the limited source of engraftable stem cells, (b) the presence of optimal time window for stem cell therapies, (c) inherited limitation of stem cells in terms of growth, trophic support, and differentiation potential, and (d) possible transplanted cell-mediated adverse effects, such as tumor formation. Here, we discuss recent advances that overcome these hurdles in adult stem cell therapy for stroke.
Collapse
Affiliation(s)
- Oh Young Bang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, Korea
| | - Eun Hee Kim
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, Korea
| | - Jae Min Cha
- Samsung Biomedical Research Institute, Samsung Advanced Institute of Technology, Samsung Electronics Co., Ltd., Seoul, Korea.,Medical Device Research Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Gyeong Joon Moon
- Translational and Stem Cell Research Laboratory on Stroke, Samsung Medical Center, Seoul, Korea.,Stem cell and Regenerative Medicine Institute, Samsung Biomedical Research Institute, Seoul, Korea
| |
Collapse
|
24
|
Cui C, Ye X, Chopp M, Venkat P, Zacharek A, Yan T, Ning R, Yu P, Cui G, Chen J. miR-145 Regulates Diabetes-Bone Marrow Stromal Cell-Induced Neurorestorative Effects in Diabetes Stroke Rats. Stem Cells Transl Med 2016; 5:1656-1667. [PMID: 27460851 PMCID: PMC5189645 DOI: 10.5966/sctm.2015-0349] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 05/13/2016] [Indexed: 01/08/2023] Open
Abstract
In rats with type 1 diabetes mellitus (T1DM) subject to stroke, the therapeutic effects and underlying mechanisms of action of bone-marrow stromal cells (BMSCs) derived from T1DM rats (DM-BMSCs) and BMSCs derived from normal rats (Nor-BMSCs) were compared. In vitro and in vivo, DM-BMSCs exhibited decreased miR-145 expression. In T1DM rats, DM-BMSC treatment significantly improved functional outcome and increased vascular and white matter remodeling. However, overexpression of miR-145 in DM-BMSCs attenuates DM-BMSC-induced neurorestorative effects in T1DM stroke rats. In rats with type 1 diabetes (T1DM), the therapeutic effects and underlying mechanisms of action of stroke treatment were compared between bone-marrow stromal cells (BMSCs) derived from T1DM rats (DM-BMSCs) and BMSCs derived from normal rats (Nor-BMSCs). The novel role of microRNA-145 (miR-145) in mediating DM-BMSC treatment-induced benefits was also investigated. T1DM rats (n = 8 per group) underwent 2 hours of middle cerebral artery occlusion (MCAo) and were treated 24 hours later with the one of the following (5 × 106 cells administered i.v.): (a) phosphate-buffered saline (PBS); (b) Nor-BMSCs; (c) DM-BMSCs; (d) DM-BMSCs with miR-145 overexpression (miR-145+/+DM-BMSCs); or (e) Nor-BMSCs with miR-145 knockdown. Evaluation of functional outcome, vascular and white-matter remodeling and microRNA expression was made, and in vitro studies were performed. In vitro, DM-BMSCs exhibited decreased miR-145 expression and increased survival compared with Nor-BMSCs. Capillary tube formation and axonal outgrowth in cultured primary cortical neurons were significantly increased by DM-BMSC-conditioned medium compared with Nor-BMSCs, and significantly decreased by miR-145+/+DM-BMSC-conditioned medium compared with DM-BMSCs. In T1DM rats in which stroke had been induced (T1DM stroke rats), DM-BMSC treatment significantly improved functional outcome, increased vascular and white matter remodeling, decreased serum miR-145 expression, and increased expression of the miR-145 target genes adenosine triphosphate-binding cassette transporter 1 (ABCA1) and insulin-like growth factor 1 receptor (IGFR1), compared with Nor-BMSCs or PBS treatment. However, miR-145+/+DM-BMSCs significantly increased serum miR-145 expression and decreased brain ABCA1 and IGFR1 expression, as well as attenuated DM-BMSC-induced neurorestorative effects in T1DM-MCAo rats. DM-BMSCs exhibited decreased miR-145 expression. In T1DM-MCAo rats, DM-BMSC treatment improved functional outcome and promoted neurorestorative effects. The miR-145/ABCA1/IGFR1 pathway may contribute to the enhanced DM-BMSCs’ functional and neurorestorative effects in T1DM stroke rats. Significance In rats with type 1 diabetes (T1DM), the therapeutic effects and underlying mechanisms of action of stroke treatment were compared between bone-marrow stromal cells (BMSCs) derived from T1DM rats (DM-BMSCs) and BMSCs derived from normal rats (Nor-BMSCs). In vitro, DM-BMSCs and derived exosomes decreased miR-145 expression and increased DM-BMSC survival, capillary tube formation, and axonal outgrowth, compared with Nor-BMSCs; these effects were decreased by DM-BMSCs in which miR-145 was overexpressed. In vivo, compared with Nor-BMSC or phosphate-buffered saline treatment, DM-BMSC treatment improved functional outcome and vascular and white matter remodeling, decreased serum miR-145 expression, and increased expression of the miR-145 target genes ABCA1 and IGFR1. microRNA-145 mediated the benefits induced by DM-BMSC treatment.
Collapse
Affiliation(s)
- Chengcheng Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, People's Republic of China
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Xinchun Ye
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Tao Yan
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Ruizhou Ning
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Peng Yu
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| |
Collapse
|
25
|
Advances in the Treatment of Ischemic Diseases by Mesenchymal Stem Cells. Stem Cells Int 2016; 2016:5896061. [PMID: 27293445 PMCID: PMC4886089 DOI: 10.1155/2016/5896061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/12/2016] [Indexed: 12/13/2022] Open
Abstract
Ischemic diseases are a group of diseases, including ischemic cerebrovascular disease, ischemic cardiomyopathy (ICM), and diabetic foot as well as other diseases which are becoming a leading cause of morbidity and mortality in the whole world. Mesenchymal stem cells (MSCs) have been used to treat a variety of ischemic diseases in animal models and clinical trials. Lots of recent publications demonstrated that MSCs therapy was safe and relieved symptoms in patients of ischemic disease. However, many factors could influence therapeutic efficacy including route of delivery, MSCs' survival and residential rate in vivo, timing of transplantation, particular microenvironment, and patient's clinical condition. In this review, the current status, therapeutic potential, and the detailed factors of MSCs-based therapeutics for ischemic cerebrovascular disease, ICM, and diabetic foot are presented and discussed. We think that MSCs transplantation would constitute an ideal option for patients with ischemic diseases.
Collapse
|
26
|
Zhilai Z, Biling M, Sujun Q, Chao D, Benchao S, Shuai H, Shun Y, Hui Z. Preconditioning in lowered oxygen enhances the therapeutic potential of human umbilical mesenchymal stem cells in a rat model of spinal cord injury. Brain Res 2016; 1642:426-435. [PMID: 27085204 DOI: 10.1016/j.brainres.2016.04.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/25/2016] [Accepted: 04/12/2016] [Indexed: 12/20/2022]
Abstract
Human umbilical cord mesenchymal stem cells (UCMSCs) have recently been shown to hold great therapeutic potential for the treatment of spinal cord injury (SCI). However, the number of engrafted cells has been shown to decrease dramatically post-transplantation. Physioxia is known to enhance the paracrine properties and immune modulation of stem cells, a notion that has been applied in many clinical settings. We therefore hypothesized that preconditioning of UCMSCs in physioxic environment would enhance the regenerative properties of these cells in the treatment of rat SCI. UCMSCs were pretreated with either atmospheric normoxia (21% O2, N-UCMSC) or physioxia (5% O2, P-UCMSC). The MSCs were characterized using flow cytometry, immunocytochemistry, and real-time polymerase chain reaction. Furthermore, 10(5) N-UCMSC or P-UCMSC were injected into the injured spinal cord immediately after SCI, and locomotor function as well as cellular, molecular and pathological changes were compared between the groups. We found that N-UCMSC and P-UCMSC displayed similar surface protein expression. P-UCMSC grew faster, while physioxia up-regulated the expression of trophic and growth factors, including hepatocyte growth factor (HGF), brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor(VEGF), in UCMSCs. Compared to N-UCMSC, treatment with P-UCMSC was associated with marked changes in the SCI environment, with a significant increase in axonal preservation and a decrease in the number of caspase-3+ cells and ED-1+ macrophages. These changes were accompanied by improved functional recovery. Thus, the present study indicated that preculturing UCMSCs under 5% lowered oxygen physioxic conditions prior to transplantation improves their therapeutic potential for the treatment of SCI in rats.
Collapse
Affiliation(s)
- Zhou Zhilai
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Mo Biling
- Department of Cardiology, Liwan Hospital, Guangzhou Medical University, China.
| | - Qiu Sujun
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Dong Chao
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Shi Benchao
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Huang Shuai
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Yao Shun
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China
| | - Zhang Hui
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye road, 510282 Guangzhou, China.
| |
Collapse
|
27
|
Kim EH, Kim DH, Kim HR, Kim SY, Kim HH, Bang OY. Stroke Serum Priming Modulates Characteristics of Mesenchymal Stromal Cells by Controlling the Expression miRNA-20a. Cell Transplant 2016; 25:1489-99. [PMID: 26762119 DOI: 10.3727/096368916x690430] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) expanded with fetal bovine serum (FBS) has some limitations, including the requirement of a long culture period to obtain a sufficient amount of stem cells. Priming of MSCs with serum from patients with ischemic stroke (stroke serum) increased the proliferation rate and the neurorestorative capacity of MSCs. We hypothesized that this novel priming method increases the proliferation rate of MSCs via the regulation of microRNAs (miRs). Thus, we investigated miR profiling in stroke serum-primed MSCs and tested whether the regulation of certain miRs may affect the proliferation rate of rat MSCs. The proliferation rate of MSCs cultured with stroke serum was higher than that of MSCs cultured with normal serum or FBS. Using miR microarray analysis, we compared the miR expression profiles between MSCs cultured in FBS and in stroke serum. Among miRs associated with cell proliferation, miR-20a was most significantly increased. Similarly, miR-20a was increased in MSCs obtained from the bone marrow of stroke rats compared with MSCs from normal rats. Furthermore, the deregulation of miR-20a by the transfection of MSCs with pre-miR-20a or anti-miR-20a was significantly correlated with the increased proliferation rate of MSCs. The overexpression of miR-20a in MSCs cultured in FBS improved the proliferation rate, while the knockdown of endogenous miR-20a decreased the proliferation rate. In addition, miR-20a promoted proliferation by suppressing the expression of p21 cyclin-dependent kinase inhibitor 1 (CDKN1A). A dual-luciferase reporter assay showed that CDKN1A is a target of miR-20a. Our findings indicate that stroke serum priming upregulated the expression of miR-20a, which promoted MSC proliferation by regulating the cell cycle inhibitor p21 CDKN1A, and suggest the possible roles of priming methods in modulating the characteristics of MSCs by controlling the expression of miR in MSCs.
Collapse
Affiliation(s)
- Eun Hee Kim
- Medical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
28
|
Zhao Q, Hu J, Xiang J, Gu Y, Jin P, Hua F, Zhang Z, Liu Y, Zan K, Zhang Z, Zu J, Yang X, Shi H, Zhu J, Xu Y, Cui G, Ye X. Intranasal administration of human umbilical cord mesenchymal stem cells-conditioned medium enhances vascular remodeling after stroke. Brain Res 2015; 1624:489-496. [PMID: 26279113 DOI: 10.1016/j.brainres.2015.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 01/24/2023]
Abstract
Stem cell-based treatments have been reported to be a potential strategy for stroke. However, tumorigenic potential and low survival rates of transplanted cells could attenuate the efficacy of the stem cell-based treatments. The application of stem cell-condition medium (CM) may be a practicable approach to conquer these limitations. In this study, we investigated whether intranasal administration of human umbilical cord mesenchymal stem cells (hUCMSCs)-CM has the therapeutic effects in rats after stroke. Adult male rats were subjected to middle cerebral artery occlusion (MCAo) and were treated by intranasal routine with or without hUCMSCs-CM (1 ml/kg/d), starting 24h after MCAo and daily for 14 days. Neurological functional tests, blood brain barrier (BBB) leakage, were measured. Angiogenesis and angiogenic factor expression were measured by immunohistochemistry, and Western blot, respectively. hUCMSCs-CM treatment of stroke by intranasal routine starting 24h after MCAo in rats significantly enhances BBB functional integrity and promotes functional outcome but does not decrease lesion volume compared to rats in DMEM/F12 medium control group and saline control group. Treatment of ischemic rats with hUCMSCs-CM by intranasal routine also significantly decreases the levels of Ang2 and increases the levels of both Ang1 and Tie2 in the ischemic brain. To take together, increased expression of Ang1 and Tie2 and decreased expression of Ang2, induced by hUCMSCs-CM treatment, contribute to vascular remodeling in the ischemic brain which plays an important role in functional outcome after stroke.
Collapse
Affiliation(s)
- Qiuchen Zhao
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jinxia Hu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jie Xiang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Yuming Gu
- Department of Interventional Radiology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Peisheng Jin
- Department of Plastic Surgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Fang Hua
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Zunsheng Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Yonghai Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Kun Zan
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Zuohui Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jie Zu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Xinxin Yang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Hongjuan Shi
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jienan Zhu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Yun Xu
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Xinchun Ye
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China.
| |
Collapse
|
29
|
Sygnecka K, Heider A, Scherf N, Alt R, Franke H, Heine C. Mesenchymal stem cells support neuronal fiber growth in an organotypic brain slice co-culture model. Stem Cells Dev 2014; 24:824-35. [PMID: 25390472 DOI: 10.1089/scd.2014.0262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been identified as promising candidates for neuroregenerative cell therapies. However, the impact of different isolation procedures on the functional and regenerative characteristics of MSC populations has not been studied thoroughly. To quantify these differences, we directly compared classically isolated bulk bone marrow-derived MSCs (bulk BM-MSCs) to the subpopulation Sca-1(+)Lin(-)CD45(-)-derived MSCs(-) (SL45-MSCs), isolated by fluorescence-activated cell sorting from bulk BM-cell suspensions. Both populations were analyzed with respect to functional readouts, that are, frequency of fibroblast colony forming units (CFU-f), general morphology, and expression of stem cell markers. The SL45-MSC population is characterized by greater morphological homogeneity, higher CFU-f frequency, and significantly increased nestin expression compared with bulk BM-MSCs. We further quantified the potential of both cell populations to enhance neuronal fiber growth, using an ex vivo model of organotypic brain slice co-cultures of the mesocortical dopaminergic projection system. The MSC populations were cultivated underneath the slice co-cultures without direct contact using a transwell system. After cultivation, the fiber density in the border region between the two brain slices was quantified. While both populations significantly enhanced fiber outgrowth as compared with controls, purified SL45-MSCs stimulated fiber growth to a larger degree. Subsequently, we analyzed the expression of different growth factors in both cell populations. The results show a significantly higher expression of brain-derived neurotrophic factor (BDNF) and basic fibroblast growth factor in the SL45-MSCs population. Altogether, we conclude that MSC preparations enriched for primary MSCs promote neuronal regeneration and axonal regrowth, more effectively than bulk BM-MSCs, an effect that may be mediated by a higher BDNF secretion.
Collapse
Affiliation(s)
- Katja Sygnecka
- 1 Translational Centre for Regenerative Medicine (TRM), University of Leipzig , Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
30
|
Qin X, Han W, Yu Z. Neuronal-like differentiation of bone marrow-derived mesenchymal stem cells induced by striatal extracts from a rat model of Parkinson's disease. Neural Regen Res 2014; 7:2673-80. [PMID: 25337113 PMCID: PMC4200735 DOI: 10.3969/j.issn.1673-5374.2012.34.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 11/27/2012] [Indexed: 01/31/2023] Open
Abstract
A rat model of Parkinson's disease was established by 6-hydroxydopamine injection into the medial forebrain bundle. Bone marrow-derived mesenchymal stem cells (BMSCs) were isolated from the femur and tibia, and were co-cultured with 10% and 60% lesioned or intact striatal extracts. The results showed that when exposed to lesioned striatal extracts, BMSCs developed bipolar or multi-polar morphologies, and there was an increase in the percentage of cells that expressed glial fibrillary acidic protein (GFAP), nestin and neuron-specific enolase (NSE). Moreover, the percentage of NSE-positive cells increased with increasing concentrations of lesioned striatal extracts. However, intact striatal extracts only increased the percentage of GFAP-positive cells. The findings suggest that striatal extracts from Parkinson's disease rats induce BMSCs to differentiate into neuronal-like cells in vitro.
Collapse
Affiliation(s)
- Xiaoling Qin
- Department of Neurology, Xuzhou Central Hospital, Xuzhou 221009, Jiangsu Province, China
| | - Wang Han
- Department of Emergency, Dongying People's Hospital, Dongying 257091, Shandong Province, China
| | - Zhigang Yu
- Department of Neurology, Xuzhou Central Hospital, Xuzhou 221009, Jiangsu Province, China
| |
Collapse
|
31
|
Yang Z, Zhu L, Li F, Wang J, Wan H, Pan Y. Bone marrow stromal cells as a therapeutic treatment for ischemic stroke. Neurosci Bull 2014; 30:524-34. [PMID: 24817388 DOI: 10.1007/s12264-013-1431-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 12/12/2013] [Indexed: 12/15/2022] Open
Abstract
Cerebral ischemia remains the most frequent cause of death and quality-of-life impairments due to neurological deficits, and accounts for the majority of total healthcare costs. However, treatments for cerebral ischemia are limited. Over the last decade, bone marrow stromal cell (BMSC) therapy has emerged as a particularly appealing option, as it is possible to help patients even when initiated days or even weeks after the ischemic insult. BMSCs are a class of multipotent, self-renewing cells that give rise to differentiated progeny when implanted into appropriate tissues. Therapeutic effects of BMSC treatment for ischemic stroke, including sensory and motor recovery, have been reported in pre-clinical studies and clinical trials. In this article, we review the recent progress in BMSC-based therapy for ischemic stroke, focusing on the route of delivery and pre-processing of BMSCs. Selecting an optimal delivery route is of particular importance. The ideal approach, as well as the least risky, for translational applications still requires further identification. Appropriate preprocessing of BMSCs or combination therapy has the benefit of achieving the maximum possible restoration. Further pre-clinical studies are required to determine the time-window for transplantation and the appropriate dosage of cells.
Collapse
Affiliation(s)
- Zizhen Yang
- Department of Neurology, First Hospital and Clinical College, Harbin Medical University, Harbin, 150001, China
| | | | | | | | | | | |
Collapse
|
32
|
Stem cell-based therapies for ischemic stroke. BIOMED RESEARCH INTERNATIONAL 2014; 2014:468748. [PMID: 24719869 PMCID: PMC3955655 DOI: 10.1155/2014/468748] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/16/2014] [Indexed: 12/16/2022]
Abstract
In recent years, stem cell-based approaches have attracted more attention from scientists and clinicians due to their possible therapeutical effect on stroke. Animal studies have demonstrated that the beneficial effects of stem cells including embryonic stem cells (ESCs), inducible pluripotent stem cells (iPSCs), neural stem cells (NSCs), and mesenchymal stem cell (MSCs) might be due to cell replacement, neuroprotection, endogenous neurogenesis, angiogenesis, and modulation on inflammation and immune response. Although several clinical studies have shown the high efficiency and safety of stem cell in stroke management, mainly MSCs, some issues regarding to cell homing, survival, tracking, safety, and optimal cell transplantation protocol, such as cell dose and time window, should be addressed. Undoubtably, stem cell-based gene therapy represents a novel potential therapeutic strategy for stroke in future.
Collapse
|
33
|
Recovery of neurological function of ischemic stroke by application of conditioned medium of bone marrow mesenchymal stem cells derived from normal and cerebral ischemia rats. J Biomed Sci 2014; 21:5. [PMID: 24447306 PMCID: PMC3922747 DOI: 10.1186/1423-0127-21-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/23/2013] [Indexed: 01/01/2023] Open
Abstract
Background Several lines of evidence have demonstrated that bone marrow-derived mesenchymal stem cells (BM-MSC) release bioactive factors and provide neuroprotection for CNS injury. However, it remains elusive whether BM-MSC derived from healthy donors or stroke patients provides equal therapeutic potential. The present work aims to characterize BM-MSC prepared from normal healthy rats (NormBM-MSC) and cerebral ischemia rats (IschBM-MSC), and examine the effects of their conditioned medium (Cm) on ischemic stroke animal model. Results Isolated NormBM-MSC or IschBM-MSC formed fibroblastic like morphology and expressed CD29, CD90 and CD44 but failed to express the hematopoietic marker CD34. The number of colony formation of BM-MSC was more abundant in IschBM-MSC than in NormBM-MSC. This is in contrast to the amount of Ficoll-fractionated mononuclear cells from normal donor and ischemic rats. The effect of cm of BM-MSC was further examined in cultures and in middle cerebral artery occlusion (MCAo) animal model. Both NormBM-MSC Cm and IschBM-MSC Cm effectively increased neuronal connection and survival in mixed neuron-glial cultures. In vivo, intravenous infusion of NormBM-MSC Cm and IschBM-MSC Cm after stroke onset remarkably improved functional recovery. Furthermore, NormBM-MSC Cm and IschBM-MSC Cm increased neurogenesis and attenuated microglia/ macrophage infiltration in MCAo rat brains. Conclusions Our data suggest equal effectiveness of BM-MSC Cm derived from ischemic animals or from a normal population. Our results thus revealed the potential of BM-MSC Cm on treatment of ischemic stroke.
Collapse
|
34
|
Shehadah A, Chen J, Pal A, He S, Zeitlin A, Cui X, Zacharek A, Cui Y, Roberts C, Lu M, Hariri R, Chopp M. Human placenta-derived adherent cell treatment of experimental stroke promotes functional recovery after stroke in young adult and older rats. PLoS One 2014; 9:e86621. [PMID: 24466174 PMCID: PMC3897748 DOI: 10.1371/journal.pone.0086621] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/16/2013] [Indexed: 12/29/2022] Open
Abstract
Background Human Placenta-Derived Adherent Cells (PDAC®) are a novel mesenchymal-like cell population derived from normal human placental tissue. PDA-001 is a clinical formulation of PDAC® developed for intravenous administration. In this study, we investigated the efficacy of PDA-001 treatment in a rat model of transient middle cerebral artery occlusion (MCAo) in young adult (2–3 month old) and older rats (10–12 months old). Methods To evaluate efficacy and determine the optimal number of transplanted cells, young adult Wistar rats were subjected to MCAo and treated 1 day post MCAo with 1×106, 4×106 or 8×106 PDA-001 cells (i.v.), vehicle or cell control. 4×106 or 8×106 PDA-001 cells were also tested in older rats after MCAo. Treatment response was evaluated using a battery of functional outcome tests, consisting of adhesive-removal test, modified Neurological Severity Score (mNSS) and foot-fault test. Young adult rats were sacrificed 56 days after MCAo, older rats were sacrificed 29 days after MCAo, and lesion volumes were measured using H&E. Immunohistochemical stainings for bromodeoxyuridine (BrdU) and von Willebrand Factor (vWF), and synaptophysin were performed. Results In young adult rats, treatment with 4×106 PDA-001 cells significantly improved functional outcome after stroke (p<0.05). In older rats, significant functional improvement was observed with PDA-001 cell therapy in both of the 4×106 and 8×106 treatment groups. Functional benefits in young adult and older rats were associated with significant increases in the number of BrdU immunoreactive endothelial cells, vascular density and perimeter in the ischemic brain, as well as significantly increased synaptophysin expression in the ischemic border zone (p<0.05). Conclusion PDA-001 treatment significantly improved functional outcome after stroke in both young adult and older rats. The neurorestorative effects induced by PDA-001 treatment may be related to increased vascular density and synaptic plasticity.
Collapse
Affiliation(s)
- Amjad Shehadah
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
- * E-mail:
| | - Ajai Pal
- Celgene Cellular Therapeutics, Warren, New Jersey, United States of America
| | - Shuyang He
- Celgene Cellular Therapeutics, Warren, New Jersey, United States of America
| | - Andrew Zeitlin
- Celgene Cellular Therapeutics, Warren, New Jersey, United States of America
| | - Xu Cui
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Yisheng Cui
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Cynthia Roberts
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Mei Lu
- Department of Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Robert Hariri
- Celgene Cellular Therapeutics, Warren, New Jersey, United States of America
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, United States of America
- Department of Physics, Oakland University, Rochester, Michigan, United States of America
| |
Collapse
|
35
|
Li Y, Liu Z, Xin H, Chopp M. The role of astrocytes in mediating exogenous cell-based restorative therapy for stroke. Glia 2013; 62:1-16. [PMID: 24272702 DOI: 10.1002/glia.22585] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 08/08/2013] [Accepted: 09/18/2013] [Indexed: 12/19/2022]
Abstract
Astrocytes have not been a major therapeutic target for the treatment of stroke, with most research emphasis on the neuron. Given the essential role that astrocytes play in maintaining physiological function of the central nervous system and the very rapid and sensitive reaction astrocytes have in response to cerebral injury or ischemic insult, we propose to replace the neurocentric view for treatment with a more nuanced astrocytic centered approach. In addition, after decades of effort in attempting to develop neuroprotective therapies, which target reduction of the ischemic lesion, there are no effective clinical treatments for stroke, aside from thrombolysis with tissue plasminogen activator, which is used in a small minority of patients. A more promising therapeutic approach, which may affect nearly all stroke patients, may be in promoting endogenous restorative mechanisms, which enhance neurological recovery. A focus of efforts in stimulating recovery post stroke is the use of exogenously administered cells. The present review focuses on the role of the astrocyte in mediating the brain network, brain plasticity, and neurological recovery post stroke. As a model to describe the interaction of a restorative cell-based therapy with astrocytes, which drives recovery from stroke, we specifically highlight the subacute treatment of stroke with multipotent mesenchymal stromal cell therapy.
Collapse
Affiliation(s)
- Yi Li
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan
| | | | | | | |
Collapse
|
36
|
Yoo J, Seo JJ, Eom JH, Hwang DY. Enhanced recovery from chronic ischemic injury by bone marrow cells in a rat model of ischemic stroke. Cell Transplant 2013; 24:167-82. [PMID: 24152766 DOI: 10.3727/096368913x674666] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Even after decades of intensive studies, therapeutic options for patients with stroke are rather limited. Thrombolytic drugs effectively treat the very acute stage of stroke, and several neuroprotectants that are designed to treat secondary injury following stroke are being tested in clinical trials. However, these pharmacological approaches primarily focus on acute stroke recovery, and few options are available for treating chronic stroke patients. In recent years, stem cell-mediated regenerative approaches have emerged as promising therapeutic strategies for treating the chronic stage of stroke. In this study, we examined whether systemically administered bone marrow cells (BMCs) could have beneficial effects in a rat model of chronic ischemia. Our transplantation experiments using BMCs obtained from ischemic donor rats showed functional and structural recovery during the chronic stage of stroke. BMC-mediated neural proliferation was prominent in the brains of rats with chronic stroke, and most of the new cells eventually became neurons instead of astrocytes. BMC-mediated enhanced neural proliferation coincided with a significant reduction (∼50%) in the number of activated microglia, which is consistent with previous reports of enhanced neural proliferation being linked to microglial inactivation. Strikingly, approximately 57% of the BMCs that infiltrated the chronic ischemic brain were CD25(+) cells, suggesting that these cells may exert the beneficial effects associated with BMC transplantation. Based on the reported anti-inflammatory role of CD25(+) regulatory T-cells in acute experimental stroke, we propose a working model delineating the positive effects of BMC transplantation during the chronic phase of stroke; infiltrating BMCs (mostly CD25(+) cells) reduce activated microglia, which leads to enhanced neural proliferation and enhanced recovery from neuronal damage in this rat model of chronic stroke. This study provides valuable insights into the effect of BMC transplantation in the chronic ischemic brain, which may lead to the development of effective therapy for chronic stroke patients who currently lack satisfactory therapeutic options.
Collapse
Affiliation(s)
- Jongman Yoo
- Department of Biomedical Science, College of Life Science, CHA University, Gyeonggido, Korea
| | | | | | | |
Collapse
|
37
|
Kim SJ, Moon GJ, Chang WH, Kim YH, Bang OY. Intravenous transplantation of mesenchymal stem cells preconditioned with early phase stroke serum: current evidence and study protocol for a randomized trial. Trials 2013; 14:317. [PMID: 24083670 PMCID: PMC4016561 DOI: 10.1186/1745-6215-14-317] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 09/12/2013] [Indexed: 12/19/2022] Open
Abstract
Background Recovery after a major stroke is usually limited, but cell therapy for patients with fixed neurologic deficits is emerging. Several recent clinical trials have investigated mesenchymal stem cell (MSC) therapy for patients with ischemic stroke. We previously reported the results of a controlled trial on the application of autologous MSCs in patients with ischemic stroke with a long-term follow-up of up to 5 years (the 'STem cell Application Researches and Trials In NeuroloGy’ (STARTING) study). The results from this pilot trial are challenging, but also raise important issues. In addition, there have been recent efforts to improve the safety and efficacy of MSC therapy for stroke. Methods and design The clinical and preclinical background and the STARTING-2 study protocol are provided. The trial is a prospective, randomized, open-label, blinded-endpoint (PROBE) clinical trial. Both acute and chronic stroke patients will be selected based on clinical and radiological features and followed for 3 months after MSC treatment. The subjects will be randomized into one of two groups: (A) a MSC group (n = 40) or (B) a control group (n = 20). Autologous MSCs will be intravenously administered after ex vivo culture expansion with autologous ischemic serum obtained as early as possible, to enhance the therapeutic efficacy (ischemic preconditioning). Objective outcome measurements will be performed using multimodal MRI and detailed functional assessments by blinded observers. Discussion This trial is the first to evaluate the efficacy of MSCs in patients with ischemic stroke. The results may provide better evidence for the effectiveness of MSC therapy in patients with ischemic stroke. Trial registration This trial was registered with ClinicalTrials.gov, number NCT01716481.
Collapse
Affiliation(s)
- Suk Jae Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, 135-710, Gangnam-gu, Seoul, South Korea.
| | | | | | | | | | | |
Collapse
|
38
|
Wan H, Li F, Zhu L, Wang J, Yang Z, Pan Y. Update on therapeutic mechanism for bone marrow stromal cells in ischemic stroke. J Mol Neurosci 2013; 52:177-85. [PMID: 24048741 DOI: 10.1007/s12031-013-0119-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/09/2013] [Indexed: 02/08/2023]
Abstract
Cerebral ischemia is a major cause of morbidity and mortality in the aged population, as well as a tremendous burden on the healthcare system. Despite timely treatment with thrombolysis and percutaneous intravascular interventions, many patients are often left with irreversible neurological deficits. Bone marrow stromal cells (BMSCs), also referred to as mesenchymal stem cells (MSCs), are a type of nonhematopoietic stem cells which exists in bone marrow mesh, with the potential to self-renew. Unlike cells in the central nervous system, BMSCs differentiate not only into mesodermal cells, but also endodermal and ectodermal cells. Moreover, it has been reported that BMSCs develop into cells with neural and vascular markers and play a role in recovery from ischemic stroke. These findings have fuelled excitement in regenerative medicine for neurological diseases, especially for ischemic stroke. There is now preclinical evidence to suggest that BMSCs grafted into the brain of ischemic models abrogate neurological deficits. Based on the overwhelming evidence from animal studies as well as in clinical trials, BMSC transplantation is considered a promising strategy for treatment of ischemic stroke. The goal of this review is to present an integrated consideration of molecular mechanisms in a chronological fashion and discuss an optimal BMSC delivery route for ischemic stroke.
Collapse
Affiliation(s)
- Huan Wan
- Department of Neurology, First Hospital and Clinical College, Harbin Medical University, Room 501, Building 3, 23 Youzheng, Harbin, 150001, China
| | | | | | | | | | | |
Collapse
|
39
|
Eckert MA, Vu Q, Xie K, Yu J, Liao W, Cramer SC, Zhao W. Evidence for high translational potential of mesenchymal stromal cell therapy to improve recovery from ischemic stroke. J Cereb Blood Flow Metab 2013; 33:1322-34. [PMID: 23756689 PMCID: PMC3764389 DOI: 10.1038/jcbfm.2013.91] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 12/27/2022]
Abstract
Although ischemic stroke is a major cause of morbidity and mortality, current therapies benefit only a small proportion of patients. Transplantation of mesenchymal stromal cells (MSC, also known as mesenchymal stem cells or multipotent stromal cells) has attracted attention as a regenerative therapy for numerous diseases, including stroke. Mesenchymal stromal cells may aid in reducing the long-term impact of stroke via multiple mechanisms that include induction of angiogenesis, promotion of neurogenesis, prevention of apoptosis, and immunomodulation. In this review, we discuss the clinical rationale of MSC for stroke therapy in the context of their emerging utility in other diseases, and their recent clinical approval for treatment of graft-versus-host disease. An analysis of preclinical studies examining the effects of MSC therapy after ischemic stroke indicates near-universal agreement that MSC have significant favorable effect on stroke recovery, across a range of doses and treatment time windows. These results are interpreted in the context of completed and ongoing human clinical trials, which provide support for MSC as a safe and potentially efficacious therapy for stroke recovery in humans. Finally, we consider principles of brain repair and manufacturing considerations that will be useful for effective translation of MSC from the bench to the bedside for stroke recovery.
Collapse
Affiliation(s)
- Mark A Eckert
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| | - Quynh Vu
- Department of Neurology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Kate Xie
- Department of Neurology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Jingxia Yu
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| | - Wenbin Liao
- Department of Pathology, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Steven C Cramer
- Departments of Neurology and Anatomy and Neurobiology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Weian Zhao
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| |
Collapse
|
40
|
Knoll A, Brockmeyer T, Chevalier R, Zscheppang K, Nielsen H, Dammann C. Adult Rat Bone Marrow-Derived Stem Cells Promote Late Fetal Type II Cell Differentiation in a Co-Culture Model. Open Respir Med J 2013; 7:46-53. [PMID: 23730368 PMCID: PMC3664445 DOI: 10.2174/1874306401307010046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 03/08/2013] [Accepted: 03/13/2013] [Indexed: 01/08/2023] Open
Abstract
Bronchopulmonary dysplasia develops in preterm infants due to a combination of lung immaturity and lung injury. Cultured pluripotent bone marrow stem cells (BMSC) are known to reduce injury and induce repair in adult and in immature lungs, possibly through paracrine secretion of soluble factors. The paracrine relationship between BMSC and primary fetal lung epithelial type II cells is unknown. We determined the effects of BMSC on type II cell and fibroblast behavior using an in vitro co-culture model. Rat BMSC were isolated and co-cultured with primary fetal E21 rat type II cells or lung fibroblasts in a Transwell® system without direct cell contact. Effects of BMSC conditioned media (CM) on type II cell and fibroblast proliferation and on type II cell surfactant phospholipid (DSPC) synthesis and mRNA expression of surfactant proteins B and C (sftpb and sftpc) were studied. We also determined the effect of fibroblast and type II cell CM on BMSC proliferation and surface marker expression. Co-culture with BMSC significantly decreased type II cell and fibroblast proliferation to 72.5% and 83.7% of controls, respectively. Type II cell DSPC synthesis was significantly increased by 21% and sftpb and sftpc mRNA expressions were significantly induced (2.1 fold and 2.4 fold, respectively). BMSC proliferation was significantly reduced during the co-culture. Flow cytometry confirmed that BMSC retained the expression of undifferentiated stem cell markers despite their exposure to fetal lung cell CM. We conclude that BMSC induce fetal type II cell differentiation through paracrine release of soluble factors. These studies provide clues for how BMSC may act in promoting alveolar repair following injury.
Collapse
Affiliation(s)
- Ab Knoll
- Hannover Medical School, Hannover, Germany ; Division of Newborn Medicine, Floating Hospital for Children at Tufts Medical Center, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
41
|
Ström JO, Ingberg E, Theodorsson A, Theodorsson E. Method parameters' impact on mortality and variability in rat stroke experiments: a meta-analysis. BMC Neurosci 2013; 14:41. [PMID: 23548160 PMCID: PMC3637133 DOI: 10.1186/1471-2202-14-41] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/22/2013] [Indexed: 12/14/2022] Open
Abstract
Background Even though more than 600 stroke treatments have been shown effective in preclinical studies, clinically proven treatment alternatives for cerebral infarction remain scarce. Amongst the reasons for the discrepancy may be methodological shortcomings, such as high mortality and outcome variability, in the preclinical studies. A common approach in animal stroke experiments is that A) focal cerebral ischemia is inflicted, B) some type of treatment is administered and C) the infarct sizes are assessed. However, within this paradigm, the researcher has to make numerous methodological decisions, including choosing rat strain and type of surgical procedure. Even though a few studies have attempted to address the questions experimentally, a lack of consensus regarding the optimal methodology remains. Methods We therefore meta-analyzed data from 502 control groups described in 346 articles to find out how rat strain, procedure for causing focal cerebral ischemia and the type of filament coating affected mortality and infarct size variability. Results The Wistar strain and intraluminal filament procedure using a silicone coated filament was found optimal in lowering infarct size variability. The direct and endothelin methods rendered lower mortality rate, whereas the embolus method increased it compared to the filament method. Conclusions The current article provides means for researchers to adjust their middle cerebral artery occlusion (MCAo) protocols to minimize infarct size variability and mortality.
Collapse
Affiliation(s)
- Jakob O Ström
- Department of Clinical and Experimental Medicine, Clinical Chemistry, Faculty of Health Sciences, Linköping University, County Council of Östergötland, Linköping, Sweden.
| | | | | | | |
Collapse
|
42
|
Efficacy of single and multiple injections of human umbilical tissue-derived cells following experimental stroke in rats. PLoS One 2013; 8:e54083. [PMID: 23342081 PMCID: PMC3544758 DOI: 10.1371/journal.pone.0054083] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 12/05/2012] [Indexed: 11/19/2022] Open
Abstract
Introduction Human umbilical tissue-derived cells (hUTC) are a promising source of cells for regenerative treatment of stroke. In this study, we tested the efficacy of hUTC in experimental stroke and whether multiple injections of hUTC provide additional therapeutic benefits as compared to a single injection. Methods Adult male Wistar rats were subjected to 2 hours of middle cerebral artery occlusion (MCAo), and randomly selected animals were injected (i.v) with 3×106 hUTC or with vehicle control (at day: 1, 1&3 or 1&7 after MCAo, n = 8–9/group). A battery of functional outcome tests was performed at days 1, 7, 14, 21, 28, 35, 42, 49, 56 and 63 after MCAo. Rats were sacrificed at 63 days after MCAo and lesion volumes were measured. To investigate the underlying mechanism of hUTC treatment of stroke, Von Willebrand Factor (vWF), and Synaptophysin immunostaining were performed. Results All hUTC treated groups, single or multiple injections, had better functional recovery compared to control (p<0.01). There was no statistically significant difference between a single and multiple injections of hUTC (p = 0.23) or between different multiple injections groups (p>0.07) in functional outcome. All hUTC treatment groups showed significant increases in Synaptophysin, vascular density and perimeter compared to the control group (p<0.05). There was no statistically significant difference between a single and multiple injections of hUTC or between the two groups of multiple injections in all immunohistochemical measurements (p>0.1). Conclusion hUTC treatment significantly improves long term functional outcome after stroke and promotes vascular density and synaptic plasticity. At the proscribed doses, multiple injections of hUTC were not superior to single injection therapy in both functional outcome and histological assessments.
Collapse
|
43
|
Gutiérrez-Fernández M, Fuentes B, Rodríguez-Frutos B, Ramos-Cejudo J, Vallejo-Cremades MT, Díez-Tejedor E. Trophic factors and cell therapy to stimulate brain repair after ischaemic stroke. J Cell Mol Med 2012; 16:2280-90. [PMID: 22452968 PMCID: PMC3823421 DOI: 10.1111/j.1582-4934.2012.01575.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 03/21/2012] [Indexed: 12/11/2022] Open
Abstract
Brain repair involves a compendium of natural mechanisms that are activated following stroke. From a therapeutic viewpoint, reparative therapies that encourage cerebral plasticity are needed. In the last years, it has been demonstrated that modulatory treatments for brain repair such as trophic factor- and stem cell-based therapies can promote neurogenesis, gliogenesis, oligodendrogenesis, synaptogenesis and angiogenesis, all of which having a beneficial impact on infarct volume, cell death and, finally, and most importantly, on the functional recovery. However, even when promising results have been obtained in a wide range of experimental animal models and conditions these preliminary results have not yet demonstrated their clinical efficacy. Here, we focus on brain repair modulatory treatments for ischaemic stroke, that use trophic factors, drugs with trophic effects and stem cell therapy. Important and still unanswered questions for translational research ranging from experimental animal models to recent and ongoing clinical trials are reviewed here.
Collapse
Affiliation(s)
- María Gutiérrez-Fernández
- Department of Neurology and Stroke Centre, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| | - Blanca Fuentes
- Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| | - Berta Rodríguez-Frutos
- Department of Neurology and Stroke Centre, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| | - Jaime Ramos-Cejudo
- Department of Neurology and Stroke Centre, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| | - María Teresa Vallejo-Cremades
- Department of Neurology and Stroke Centre, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| | - Exuperio Díez-Tejedor
- Department of Neurology and Stroke Centre, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
- Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital Neuroscience Area of IdiPAZ (Health Research Institute) Autónoma University of MadridMadrid, Spain
| |
Collapse
|
44
|
Yang B, Xi X, Aronowski J, Savitz SI. Ischemic stroke may activate bone marrow mononuclear cells to enhance recovery after stroke. Stem Cells Dev 2012; 21:3332-40. [PMID: 22731389 DOI: 10.1089/scd.2012.0037] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Bone marrow-derived mononuclear cells (MNCs) enhance recovery in rodent stroke models. Since stroke activates the bone marrow, there may be biological differences of autologous MNCs derived poststroke compared with the prestroke setting. We analyzed MNCs harvested from the same Long Evans rats 1 day before and 1 day after ischemic stroke or sham stroke. Stroke was induced by suture occlusion of the middle cerebral artery for 90 min. MNCs were characterized by flow cytometry to identify differences in the percentages of different cell subpopulations. MNCs were also placed in culture and cytokines were measured in the media. In separate experiments, Long Evans rats received 24 h after stroke an intracarotid injection of saline or autologous MNCs, prepared from the same animal, either 1 day before or 1 day after stroke. The rats were then followed on the cylinder and corner tests for 28 days. In poststroke MNCs compared with prestroke MNCs, there was a significant reduction in T and mesenchymal stem cells and a significant increase in CD34+ and natural killer cells. Postsham MNCs showed an elevation in CD11b and CD45R cells compared with presham MNCs. The concentrations of IL-10, IL-6, MCP-1, vascular endothelial growth factor (VEGF), and tumor necrosis factor-α were significantly increased in poststroke MNCs compared with prestroke MNCs. Postsham MNCs showed a decrease in VEGF. Poststroke MNCs in comparison with prestroke MNCs led to a greater recovery on neurological testing and reduced lesion size. Autologous MNCs exert different biological responses when derived from the poststroke setting compared with normal animals.
Collapse
Affiliation(s)
- Bing Yang
- Department of Neurology, University of Texas Medical School at Houston, UT-Health, Houston, TX, USA
| | | | | | | |
Collapse
|
45
|
Kim SJ, Moon GJ, Cho YH, Kang HY, Hyung NK, Kim D, Lee JH, Nam JY, Bang OY. Circulating mesenchymal stem cells microparticles in patients with cerebrovascular disease. PLoS One 2012; 7:e37036. [PMID: 22615882 PMCID: PMC3352849 DOI: 10.1371/journal.pone.0037036] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 04/12/2012] [Indexed: 01/01/2023] Open
Abstract
Preclinical and clinical studies have shown that the application of CD105+ mesenchymal stem cells (MSCs) is feasible and may lead to recovery after stroke. In addition, circulating microparticles are reportedly functional in various disease conditions. We tested the levels of circulating CD105+ microparticles in patients with acute ischemic stroke. The expression of CD105 (a surface marker of MSCs) and CXCR4 (a CXC chemokine receptor for MSC homing) on circulating microparticles was evaluated by flow cytometry of samples from 111 patients and 50 healthy subjects. The percentage of apoptotic CD105 microparticles was determined based on annexin V (AV) expression. The relationship between serum levels of CD105+/AV− microparticles, stromal cells derived factor-1α (SDF-1α), and the extensiveness of cerebral infarcts was also evaluated. CD105+/AV− microparticles were higher in stroke patients than control subjects. Correlation analysis showed that the levels of CD105+/AV− microparticles increased as the baseline stroke severity increased. Multivariate testing showed that the initial severity of stroke was independently associated with circulating CD105+/AV− microparticles (OR, 1.103 for 1 point increase in the NIHSS score on admission; 95% CI, 1.032–1.178) after adjusting for other variables. The levels of CD105+/CXCR4+/AV− microparticles were also increased in patients with severe disability (r = 0.192, p = 0.046 for NIHSS score on admission), but were decreased with time after stroke onset (r = −0.204, p = 0.036). Risk factor profiles were not associated with the levels of circulating microparticles or SDF-1α. In conclusion, our data showed that stroke triggers the mobilization of MSC-derived microparticles, especially in patients with extensive ischemic stroke.
Collapse
Affiliation(s)
- Suk Jae Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Gyeong Joon Moon
- Clinical Research Center, Samsung Biomedical Research Institute, Seoul, South Korea
| | - Yeon Hee Cho
- Clinical Research Center, Samsung Biomedical Research Institute, Seoul, South Korea
| | - Ho Young Kang
- Clinical Research Center, Samsung Biomedical Research Institute, Seoul, South Korea
| | - Na Kyum Hyung
- Clinical Trial Center, Samsung Medical Center, Seoul, South Korea
| | - Donghee Kim
- Clinical Research Center, Samsung Biomedical Research Institute, Seoul, South Korea
- Samsung Advanced Institute for Health Sciences and Technology, Seoul, South Korea
| | - Ji Hyun Lee
- Clinical Research Center, Samsung Biomedical Research Institute, Seoul, South Korea
- Samsung Advanced Institute for Health Sciences and Technology, Seoul, South Korea
| | - Ji Yoon Nam
- Clinical Research Center, Samsung Biomedical Research Institute, Seoul, South Korea
| | - Oh Young Bang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Samsung Advanced Institute for Health Sciences and Technology, Seoul, South Korea
- * E-mail:
| |
Collapse
|
46
|
Transplantation of bone marrow mononuclear cells decreases seizure incidence, mitigates neuronal loss and modulates pro-inflammatory cytokine production in epileptic rats. Neurobiol Dis 2012; 46:302-13. [DOI: 10.1016/j.nbd.2011.12.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 10/25/2011] [Accepted: 12/04/2011] [Indexed: 11/23/2022] Open
|
47
|
Wei L, Fraser JL, Lu ZY, Hu X, Yu SP. Transplantation of hypoxia preconditioned bone marrow mesenchymal stem cells enhances angiogenesis and neurogenesis after cerebral ischemia in rats. Neurobiol Dis 2012; 46:635-45. [PMID: 22426403 DOI: 10.1016/j.nbd.2012.03.002] [Citation(s) in RCA: 273] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 02/16/2012] [Accepted: 03/01/2012] [Indexed: 12/16/2022] Open
Abstract
Hypoxic preconditioning of stem cells and neural progenitor cells has been tested for promoting cell survival after transplantation. The present investigation examined the hypothesis that hypoxic preconditioning of bone marrow mesenchymal stem cells (BMSCs) could not only enhance their survival but also reinforce regenerative properties of these cells. BMSCs from eGFP engineered rats or pre-labeled with BrdU were pre-treated with normoxia (20% O(2), N-BMSCs) or sub-lethal hypoxia (0.5% O(2). H-BMSCs). The hypoxia exposure up-regulated HIF-1α and trophic/growth factors in BMSCs, including brain-derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF) and its receptor FIK-1, erythropoietin (EPO) and its receptor EPOR, stromal derived factor-1 (SDF-1) and its CXC chemokine receptor 4 (CXCR4). Meanwhile, many pro-inflammatory cytokines/chemokines were down-regulated in H-BMSCs. N-BMSCs or H-BMSCs were intravenously injected into adult rats 24h after 90-min middle cerebral artery occlusion. Comparing to N-BMSCs, transplantation of H-BMSCs showed greater effect of suppressing microglia activity in the brain. Significantly more NeuN-positive and Glut1-positive cells were seen in the ischemic core and peri-infarct regions of the animals received H-BMSC transplantation than that received N-BMSCs. Some NeuN-positive and Glut-1-positive cells showed eGFP or BrdU immunoflourescent reactivity, suggesting differentiation from exogenous BMSCs into neuronal and vascular endothelial cells. In Rotarod test performed 15days after stroke, animals received H-BMSCs showed better locomotion recovery compared with stroke control and N-BMSC groups. We suggest that hypoxic preconditioning of transplanted cells is an effective means of promoting their regenerative capability and therapeutic potential for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
48
|
Wang Y, Geng T, Ni A, Yin H, Han B. Effects of transplanted GDNF gene modified marrow stromal cells on focal cerebral ischemia in rats. Front Integr Neurosci 2011; 5:89. [PMID: 22363270 PMCID: PMC3277278 DOI: 10.3389/fnint.2011.00089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 12/07/2011] [Indexed: 11/26/2022] Open
Abstract
Objective: To evaluate the therapeutic effect of transplanted glial cell derived neurotrophic factor (GDNF) modified marrow stromal cells (MSCs) on an experimental ischemic brain injury based on the behavioral, morphological, and immunohistochemical observations. Methods: The MSCs from four-week newborn rats were cultured in vitro. The cerebral ischemia and reperfusion model was established in adult Sprague–Dawley (SD) rats by using the suture method. Three days after model establishment, the animals were injected with prepared MSCs via their caudal veins. The animals were then divided into a sham-operation group, ischemia group, MSCs transplantation group, or GDNF+ MSCs transplantation group and were scored for their neurobehavioral manifestations at 3, 14, and 28 days after the transplantation was performed. At this time, the survival condition of intracerebral transplanted cells was measured by laser confocal microscopy while the effect of transplantation on the Generic Digital Beam Former (GDNF) expression in the ischemic brain tissue was evaluated. Results: The MSCs cells transfected with GDNF gene were characterized by green fluorescence. Three days after the transplantation, the animals that underwent the cell transplantation showed significantly better behavioral data than the controls. Fourteen days after transplantation, the animals transplanted with GDNF gene modified MSCs were better than those transplanted with common MSCs. As compared with common MSCs transplantation, GDNF+MSCs transplantation was significantly more effective in reducing apoptotic cell volume and enhancing Bcl-2 expression, which was favorable for the ischemic brain injury. Conclusions: Transplanted GDNF modified MSCs can improve the nervous function and have a protective effect on the ischemic brain injury through reducing apoptotic cell volume and enhancing the expression of anti-apoptotic gene Bcl-2.
Collapse
Affiliation(s)
- Yunliang Wang
- Department of Neurology, No. 148 Hospital of PLA, Zibo Shandong, China
| | | | | | | | | |
Collapse
|
49
|
Mochizuki N, Moriyama Y, Takagi N, Takeo S, Tanonaka K. Intravenous injection of neural progenitor cells improves cerebral ischemia-induced learning dysfunction. Biol Pharm Bull 2011; 34:260-5. [PMID: 21415538 DOI: 10.1248/bpb.34.260] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The ability of stem cells to enhance neurological recovery seen after cerebral ischemia has been reported. However, it remains to be clarified whether neural progenitor cells (NPCs) improve cerebral ischemia-induced learning dysfunction. We found in an earlier study that the direct injection of NPCs into the hippocampus prevents spatial learning dysfunction after cerebral ischemia. As the intravascular injection of cells represents a minimally invasive therapeutic approach, we sought to determine whether the intravenous injection of NPCs also would improve ischemia-induced spatial learning dysfunction. Cerebral ischemia was produced by the injection of 700 microspheres into the right hemisphere of rats. The injection of NPCs via a femoral vein on day 7 after the induction of ischemia improved the modified neurological severity score and reduced the prolongation of the escape latency seen in the water maze task on days 12-28 after cerebral ischemia. The intravenous injection of NPCs on day 7 did not affect the viable area of the ipsilateral hemisphere on day 28 compared with that of the non-treated ischemic rats. Furthermore, the NPCs injected via the vein were detected in the ipsilateral hemisphere; and they expressed brain-derived neurotrophic factor (BDNF) on day 28. The decrease in the BDNF level in the ipsilateral hemisphere was also inhibited by the injection of NPCs. These results suggest that the NPCs injected via the vein after cerebral ischemia improved spatial learning dysfunction, but without having any restorative effect on the damaged areas, possibly by acting as a source of neurotrophic factors.
Collapse
Affiliation(s)
- Nobuyuki Mochizuki
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432–1Horinouchi, Hachioji, Tokyo 192–0392, Japan
| | | | | | | | | |
Collapse
|
50
|
Font MA, Arboix A, Krupinski J. Angiogenesis, neurogenesis and neuroplasticity in ischemic stroke. Curr Cardiol Rev 2011; 6:238-44. [PMID: 21804783 PMCID: PMC2994116 DOI: 10.2174/157340310791658802] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2010] [Revised: 04/10/2010] [Accepted: 05/25/2010] [Indexed: 01/10/2023] Open
Abstract
Only very little is know about the neurovascular niche after cardioembolic stroke. Three processes implicated in neurorepair: angiogenesis, neurogenesis and synaptic plasticity, would be naturally produced in adult brains, but also could be stimulated through endogen neurorepair phenomena. Angiogenesis stimulation generates new vessels with the aim to increase collateral circulation. Neurogenesis is controlled by intrinsic genetic mechanisms and growth factors but also ambiental factors are important. The leading process of the migrating neural progenitor cells (NPCs) is closely associated with blood vessels, suggesting that this interaction provides directional guidance to the NPCs. These findings suggest that blood vessels play an important role as a scaffold for NPCs migration toward the damaged brain region. DNA microarray technology and blood genomic profiling in human stroke provided tools to investigate the expression of thousands of genes. Critical comparison of gene expression profiles after stroke in humans with those in animal models should lead to a better understanding of the pathophysiology of brain ischaemia. Probably the most important part of early recovery after stroke is limited capacity of penumbra/infarct neurones to recover. It became more clear in the last years, that penumbra is not just passively dying over time but it is also actively recovering. This initial plasticity in majority contributes towards later neurogenesis, angiogenesis and final recovery. Penumbra is a principal target in acute phase of stroke. Thus, the origin of newly formed vessels and the pathogenic role of neovascularization and neurogenesis are important unresolved issues in our understanding of the mechanisms after stroke. Biomaterials for promoting brain protection, repair and regeneration are new hot target. Recently developed biomaterials can enable and increase the target delivery of drugs or therapeutic proteins to the brain, allow cell or tissue transplants to be effectively delivered to the brain and help to rebuild damaged circuits. These new approaches are gaining clear importance because nanotechnology allows better control over material-cell interactions that induce specific developmental processes and cellular responses including differentiation, migration and outgrowth.
Collapse
|